Mutated anti-TNFα antibodies and methods of their use

- AbbVie, Inc.

The present invention is directed to modified antibodies, including anti-TNFα antibodies, in which C-terminal amino acids of heavy chain sequences are modified from a native sequence of proline-glycine-lysine (“PGK”) to one that includes a proline positioned between the glycine and lysine, resulting in a C-terminal sequence of proline-glycine-proline-lycine (“PGPK”). The invention further provides methods of producing and using such antibodies.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description
RELATED APPLICATION INFORMATION

This application claims priority to U.S. Provisional Application No. 61/784,430, filed on Mar. 14, 2013, and U.S. Provisional Application No. 61/892,710, filed on Oct. 18, 2013, the entire contents of each of which are expressly incorporated herein by reference.

BACKGROUND OF THE INVENTION

Large-scale production of antibodies for biopharmaceutical applications involves the use of cell cultures that are known to produce antibodies, and antigen-binding portions thereof, exhibiting varying levels of heterogeneity. This heterogeneity may comprise isoforms of the antibody wherein the difference between the isoforms is dependent on charge. Some of these charge variants (or isoforms) may lead to decreased product efficacy and stability. Alternatively, adding a basic overall charge to an antibody may contribute positive attributes to the antibody such as increased tissue penetration.

One source of antibody heterogeneity involves C-terminal lysine residues, such as those typically found on the heavy chains of antibody molecules, which can be lost during both the purification process and/or storage of the final composition, resulting in compositions comprising antibody species that can vary at their C-terminus as to whether a lysine residue is present. For example, C-terminal lysines can potentially be present on both the heavy chains of an antibody (Lys 2), on either one of the heavy chains (Lys 1), or neither of them (Lys 0). Since lysine carries a positive charge, antibodies lacking the basic C-terminal lysine(s) differ in their charge state from ones that contain the lysine, so that the distribution of lysine variants (% Lys 0, % Lys 1, % Lys 2 of the total Lysine Sum) can be detected by ion-exchange chromatographic methods, for example, using a ProPac WCX-10 Weak Cation-Exchange column for the high-resolution separation of protein isoforms (Dionex, Calif.), and subsequently quantified.

The development of compositions comprising antibodies, or antigen-binding portions thereof, with either higher or lower levels of lysine variants to increase efficacy and stability of antibody products is an important, to date unmet, need in the biopharmaceutical industry.

SUMMARY OF THE INVENTION

The present invention provides antibodies, and antigen-binding portions thereof, comprising mutations in their C-terminal residues that decrease C-terminal processing of lysine residues, thereby leading to more efficacious and stable antibody compositions having one or more terminal lysine residues. The present invention also provides compositions comprising antibodies, and antigen-binding portions thereof, with decreased levels of C-terminal processing of lysines.

Accordingly, in one aspect, the invention provides an antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9). In one embodiment, the antibody, or antigen-binding portion thereof, is a human antibody, or antigen-binding portion thereof. In one embodiment, the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof. In another embodiment, the antibody, or antigen-binding portion thereof, comprises a light chain variable region (LCVR) having a CDR1 domain comprising the amino acid sequence of SEQ ID NO:7, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:5, and a CDR3 domain comprising the amino acid sequence of SEQ ID NO:3; and a heavy chain variable region (HCVR) having a CDR1 domain comprising the amino acid sequence of SEQ ID NO:8, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:6, and a CDR3 domain comprising the amino acid sequence of SEQ ID NO:4. In another embodiment, the antibody, or antigen-binding portion thereof, comprises a LCVR comprising the amino acid sequence set forth in SEQ ID NO:1 and a HCVR comprising the amino acid sequence set forth in SEQ ID NO:2. In one embodiment, the antibody, or antigen-binding portion thereof, comprises adalimumab, or an antigen-binding portion thereof. In another embodiment, the antibody, or antigen-binding portion thereof, is an IgG1, an IgG2, an IgG3, or an IgG4 antibody, or antigen-binding portion thereof. In one embodiment, the antibody, or antigen-binding portion thereof, is an IgG1 antibody, or antigen-binding portion thereof.

In one embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, is resistant to C-terminal processing by a carboxypeptidase. In one embodiment, the antibody, or antigen-binding portion thereof, exhibits no C-terminal processing by a carboxypeptidase. In one embodiment, the carboxypeptidase is carboxypeptidase B. In another embodiment, the carboxypeptidase is carboxypeptidase U.

In one embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits increased tissue, e.g., cartilage, penetration as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In one embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits increased TNFα affinity as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In another embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits reduced tissue, e.g., cartilage, destruction as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In another embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits reduced bone erosion, reduced synovial proliferation, reduced cell infiltration, reduced chondrocyte death, or reduced proteoglycan loss as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In another embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits increased protection against the development of arthritic scores or increased protection against the development of histopathology scores as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10) when administered to an animal model of arthritis.

In another embodiment, the antibody, or antigen-binding portion thereof, dissociates from human TNFα with a Kd of about 1×10−8 M or less and a Koff rate constant of 1×10−3 s−1 or less.

In another aspect, the invention provides a composition comprising an antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, wherein the composition comprises less than about 50% lysine variant species that lack a C-terminal lysine (Lys 0). In one embodiment, the composition comprises less than about 25% lysine variant species that have one C-terminal lysine (Lys 1). In another embodiment, the composition comprises at least about 70% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 80% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 90% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 95% lysine variant species that have two C-terminal lysines (Lys 2).

In one embodiment, the composition comprises less than about 10% acidic species, wherein the acidic species comprise a first acidic species region (AR1) and a second acidic species region (AR2). In one embodiment, the composition comprises about 3% acidic species. In another embodiment, the composition comprises less than about 1% AR1. In another embodiment, the composition comprises about 0% AR1. In another embodiment, the composition comprises less than about 4% AR2. In another embodiment, the composition comprises about 3% AR2. In another embodiment, the composition comprises about 0% AR1 and about 3% AR2.

In another aspect, the invention provides a composition comprising an antibody, or antigen-binding portion thereof, wherein the composition comprises at least about 70% lysine variant species that have two C-terminal lysines (Lys 2). In one embodiment, the composition comprises at least about 75% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 80% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 85% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 90% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 100% lysine variant species that have two C-terminal lysines (Lys 2).

In another embodiment, the composition comprises less than about 10% acidic species, wherein the acidic species comprise a first acidic species region (AR1) and a second acidic species region (AR2). In one embodiment, the composition comprises about 3% acidic species. In another embodiment, the composition comprises less than about 1% AR1. In one embodiment, the composition comprises about 0% AR1. In another embodiment, the composition comprises less than about 4% AR2. In another embodiment, the composition comprises about 3% AR2. In one embodiment, the composition comprises about 0% AR1 and about 3% AR2.

In one embodiment, the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof. In one embodiment, the antibody, or antigen-binding portion thereof, comprises a light chain variable region (LCVR) having a CDR1 domain comprising the amino acid sequence of SEQ ID NO:7, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:5, and a CDR3 domain comprising the amino acid sequence of SEQ ID NO:3; and a heavy chain variable region (HCVR) having a CDR1 domain comprising the amino acid sequence of SEQ ID NO:8, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:6, and a CDR3 domain comprising the amino acid sequence of SEQ ID NO:4. In one embodiment, the antibody, or antigen-binding portion thereof, comprises a LCVR comprising the amino acid sequence set forth in SEQ ID NO:1 and a HCVR comprising the amino acid sequence set forth in SEQ ID NO:2. In another embodiment, the antibody, or antigen-binding portion thereof, comprises adalimumab, or an antigen-binding portion thereof.

In one embodiment, the antibody, or antigen-binding portion thereof, is resistant to C-terminal processing by a carboxypeptidase. In one embodiment, the antibody, or antigen-binding portion thereof, exhibits no C-terminal processing by a carboxypeptidase. In one embodiment, the carboxypeptidase is carboxypeptidase B. In another embodiment, the carboxypeptidase is carboxypeptidase U.

In one embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits increased cartilage tissue penetration, increased TNFα affinity, reduced cartilage destruction, reduced bone erosion, or reduced synovial proliferation as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In another embodiment, the antibody, e.g., an IgG1 antibody, or antigen-binding portion thereof, exhibits reduced cell infiltration, reduced chondrocyte death, or reduced proteoglycan loss as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10).

In another embodiment, the antibody, or antigen-binding portion thereof, exhibits increased protection against the development of arthritic scores as compared to an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (PGK) (SEQ ID NO:10) when administered to an animal model of arthritis.

In one embodiment, the antibody, or antigen-binding portion thereof, dissociates from human TNFα with a Kd of about 1×10−8 M or less and a Koff rate constant of 1×10−3 s−1 or less.

In another aspect, the invention provides a nucleic acid molecule encoding an antibody, or antigen-binding portion thereof, of the invention. In another aspect, the invention provides a vector comprising the nucleic acid molecule encoding an antibody, or antigen-binding portion thereof, of the invention. In another aspect, the invention provides a host cell comprising the vector.

In another aspect, the invention provides a composition comprising an antibody, or antigen-binding portion thereof, of the invention, and a pharmaceutically acceptable carrier.

In another aspect, the invention provides a kit comprising an antibody, or antigen-binding portion thereof, of the invention, and instructions for use.

In another aspect, the invention provides a method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering a therapeutically effective amount of an antibody, or antigen-binding portion thereof, of the invention, or a composition of the invention to the subject, thereby treating the TNFα-associated disease or disorder. In one embodiment, the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease and plaque psoriasis. In one embodiment, the disorder in which TNFα activity is detrimental is selected from the group consisting of active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

In another aspect, the invention provides a method of modifying an antibody, or antigen-binding portion thereof, to make it resistant to C-terminal processing by a carboxypeptidase, the method comprising modifying the three C-terminal amino acids of the heavy chain sequence, proline-glycine-lysine (“PGK”) (SEQ ID NO:10), of the antibody, or antigen-binding portion thereof, to introduce a proline between glycine and lysine, thereby producing an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (“PGPK”) (SEQ ID NO:9) which is resistant to C-terminal processing by a carboxypeptidase.

In another aspect, the invention provides a method of increasing the cartilage tissue penetration of an antibody, or antigen-binding portion thereof, the method comprising modifying the three C-terminal amino acids of the heavy chain sequence, proline-glycine-lysine (SEQ ID NO:10), of the antibody, or antigen-binding portion thereof, to introduce a proline between glycine and lysine, thereby producing an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (“PGPK”) (SEQ ID NO:9) which exhibits increased cartilage tissue penetration.

Other features and advantages of the invention will be apparent from the following detailed description and claims.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 depicts a WCX-10 chromatogram of a sample of Adalimumab indicating the presence of particular charge variants including Lys 0, Lys 1, and Lys 2.

FIG. 2 depicts a subsequent analysis of the WCX-purified material indicating that the individual charge variants are resolvable from one another.

FIG. 3 depicts the results of incubation of Adalimumab with or without carboxypeptidase B. In the presence of carboxypeptidase B the C-terminal lysines are cleaved leaving only Lys 0 species (with and without a particular glycol-modification (Gal)).

FIG. 4 depicts the results of incubating a modified anti-TNFα antibody with and without carboxypeptidase B. Both the treated and untreated samples display a homogenous population with the presence of an unprocessed GPK c-terminus.

FIG. 5 depicts the results of incubation of a modified anti-TNFα antibody in three different plasma samples. The modified anti-TNFα antibody retains both C-terminal lysines in all conditions.

FIG. 6 depicts the results of incubation of Adalimumab in the three different plasma samples. The antibody only retains C-terminal lysine residues in human plasma with citrate used as an anticoagulent. Without being bound by theory, the citrate presumably chelates the active site metal cation and inhibits the carboxypeptidase U activity.

FIG. 7 depicts intact LC/MS analysis of a modified anti-TNFα antibody obtained from terminal mouse bleed of an animal dosed at 5 mg/Kg. The recovered Mab has an intact GPK motif on both heavy chains.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides antibodies, and antigen-binding portions thereof, comprising mutations in their C-terminal residues that decrease C-terminal processing of lysine residues, thereby leading to more efficacious and stable antibody compositions. The present invention also provides compositions comprising antibodies, and antigen-binding portions thereof, with decreased levels of C-terminal processing of lysines.

The present invention is based, at least in part, on the discovery that the C-terminal lysines of antibodies in pharmaceutical compositions can be lost during both the purification process and/or storage of the final composition, resulting in compositions comprising individual antibody species that can vary at their C-terminus as to whether a lysine residue is present. Moreover, terminal lysines can be cleaved by enzymes, such as a carboxypeptidase, in vivo. The inventors of the instant application have surprisingly discovered that modifying an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (“PGK”) by inserting a proline between the glycine and the lysine (proline-glycine-proline-lysine, or “PGPK”) prevents the C-terminal processing of the antibody heavy chain and leads to more efficacious and stable antibody compositions.

In order that the present invention may be more readily understood, certain terms are first defined.

The term “antibody”, as used herein, broadly refers to any immunoglobulin (Ig) molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains, or any functional fragment, mutant, variant, or derivative thereof, which retains the essential epitope binding features of an Ig molecule. Such mutant, variant, or derivative antibody formats are known in the art and non-limiting embodiments of which are discussed herein.

In a full-length antibody, each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. The present invention is particularly useful for IgG1 antibodies.

As used herein, the term “adalimumab”, also known by its trade name HUMIRA® (AbbVie) refers to a human IgG1 antibody that binds human tumor necrosis factor α (TNFα). In general, the heavy chain constant domain 2 (CH2) of the adalimumab IgG-Fc region is glycosylated through covalent attachment of oligosaccharide at asparagine 297 (Asn-297). The light chain variable region of adalimumab is provided herein as SEQ ID NO:1, and the heavy chain variable region of adalimumab is provided herein as SEQ ID NO:2. Adalimumab comprises a light chain variable region comprising a CDR1 of SEQ ID NO:7, a CDR2 of SEQ ID NO:5, and a CDR3 of SEQ ID NO:3. Adalimumab comprises a heavy chain variable region comprising a CDR1 of SEQ ID NO:8, a CDR2 of SEQ ID NO:6 and CDR3 of SEQ ID NO:4. The light chain of adalimumab is provided herein as SEQ ID NO:13, and the heavy chain of adalimumab is provided herein as SEQ ID NO:14. Adalimumab is described in U.S. Pat. Nos. 6,090,382; 6,258,562; 6,509,015; 7,223,394; 7,541,031; 7,588,761; 7,863,426; 7,919,264; 8,197,813; 8,206,714; 8,216,583; 8,420,081; 8,092,998; 8,093,045; 8,187,836; 8,372,400; 8,034,906; 8,436,149; 8,231,876; 8,414,894; 8,372,401, and PCT Publication No. WO2012/065072, the entire contents of each which are expressly incorporated herein by reference in their entireties. Adalimumab is also described in “Highlights of Prescribing Information” for HUMIRA® (adalimumab) Injection (Revised January 2008).

Weak cation-exchange chromatography (WCX) analysis of adalimumab has shown that it has three main basic charge variants (i.e., Lys 0, Lys 1, and Lys 2). These variants, or charged isomers, are the result of incomplete post-translational cleavage of the C-terminal lysine residues on the heavy chains of the antibody. In addition to the lysine variants, the WCX-10 analysis measures the presence acidic species. These acidic species regions (i.e., acidic species), AR1 and AR2, are classified as product-related impurities that are relatively acidic when compared to the lysine variants and elute before the Lys 0 peak in the chromatogram (see, for example, FIG. 1).

As used herein, the term “lysine variant species” refers to an antibody, or antigen-binding portion thereof, comprising heavy chains with either zero, one or two C-terminal lysines. For example, the “Lys 0” variant comprises an antibody, or antigen-binding portion thereof, with heavy chains that do not comprise a C-terminal lysine. The “Lys 1” variant comprises an antibody, or antigen-binding portion thereof, with one heavy chain that comprises a C-terminal lysine. The “Lys 2” variant comprises an antibody, or antigen-binding portion thereof, with both heavy chains comprising a C-terminal lysine. Lysine variants can be detected by weak cation exchange chromatography, for example, WCX, of the expression product of a host cell expressing the antibody, or antigen-binding portion thereof. For example, but not by way of limitation, FIG. 2 depicts WCX analysis of adalimumab wherein the three lysine variants, as well as two acidic species, are resolved from each other.

A composition of the invention may comprise more than one lysine variant species of an antibody, or antigen-binding portion thereof. For example, in one embodiment, the composition may comprise a Lys 2 variant of an antibody, or antigen-binding portion thereof. The composition may comprise a Lys 1 variant of an antibody, or antigen-binding portion thereof. The composition may comprise a Lys 0 variant of an antibody, or antigen-binding portion thereof. In another embodiment, the composition may comprise both Lys 1 and Lys 2, or Lys 1 and Lys 0, or Lys 2 and Lys 0 variants of an antibody, or antigen-binding portion thereof. In another embodiment, the composition may comprise all three lysine variant species, i.e., Lys 0, Lys 1 and Lys 2, of an antibody, or antigen-binding portion thereof.

As used herein, the phrases “antibody resistant to C-terminal processing” or “antibody resistant to C-terminal processing by a carboxypeptidase” refer to an antibody, or antigen-binding portion thereof, that is resistant to processing of the C-terminus of its heavy chains by a carboxypeptidase enzyme, e.g., carboxypeptidase B or carboxypeptidase U. An “antibody resistant to C-terminal processing” exhibits decreased removal of a C-terminal lysine of its heavy chains by a carboxypeptidase enzyme, e.g., carboxypeptidase B or carboxypeptidase U. The antibody, or antigen-binding portion thereof, may be modified as described herein to exhibit decreased removal of a C-terminal lysine as compared to an antibody, or antigen-binding portion thereof, that has not been modified. In one embodiment, the antibody, or antigen-binding portion thereof, retains both C-terminal lysines (“Lys 2”) and, thus, exhibits no removal (i.e., exhibits no C-terminal processing) of the C-terminal lysines of the heavy chains by a carboxypeptidase. C-terminal processing by a carboxypeptidase may be measured using assays that are well-known in the art including, but not limited to, the peptidase assays described in the Examples section below.

As used herein, the term “carboxypeptidase” refers to a protease enzyme that hydrolyzes a peptide bond at the carboxy-terminal (“C-terminal”) region of a protein or antibody. Carboxypeptidases are well-known in the art and are involved in post-translational modification of proteins. Specifically, “carboxypeptidase B” (EC 3.4.17.2) refers to a carboxypeptidase that preferentially cleaves positively charged, or basic, amino acids, such as arginine and lysine from the c-terminus of proteins and antibodies. “Carboxypeptidase U” or “unstable carboxypeptidase” (EC 3.4.17.20) refers to a carboxypeptidase that is activated by thrombin or plasmin during clotting.

The term “modify”, “modifying” or “modified,” as used herein, is intended to refer to changing one or more amino acids in an antibody, or antigen-binding portion thereof. The change can be produced by adding, substituting or deleting an amino acid at one or more positions. The change can be produced using standard techniques known in the art and described in more detail herein, such as PCR mutagenesis and site-directed mutagenesis. In one embodiment, of the invention, the C-terminal three amino acids of the heavy chain sequences of an antibody, or antigen-binding portion thereof, are modified from the native sequence of proline-glycine-lysine (“PGK”) (SEQ ID NO:10) to include a proline between the glycine and lysine, resulting in a C-terminal sequence of proline-glycine-proline-lysine (“PGPK”) (SEQ ID NO:9). For example, the C-terminal three amino acids of the heavy chain sequence of adalimumab (SEQ ID NO:14) can be modified from the native sequence of proline-glycine-lysine (“PGK”) (see, e.g., SEQ ID NO:14) to include a proline between the glycine and lysine, resulting in a C-terminal sequence of proline-glycine-proline-lysine (“PGPK”) (see, e.g., SEQ ID NO:15).

As used herein, the phrase “increased cartilage tissue penetration” refers to the property of an antibody, or antigen-binding portion thereof, of the invention showing increased penetration of cartilage tissue. This property can be measured or determined by, for example, using an in vitro or an in vivo cartilage model. In one embodiment, an antibody, or antigen-binding portion thereof, retains both C-terminal lysines (“Lys 2”) and exhibits increased cartilage penetration as compared to an antibody, or antigen-binding portion thereof, having only one C-terminal lysine (“Lys 1”) or no C-terminal lysines (“Lys 0”). In one embodiment, the antibody, or antigen-binding portion thereof, has been modified to exhibit increased cartilage penetration as compared to an antibody, or antigen-binding portion thereof, that has not been modified. Cartilage penetration can be measured using assays that are well-known in the art including, but not limited to, the assays described in the Examples section below.

As used herein, the terms “acidic species”, “acidic species regions” and “AR,” refer to the variants of a protein, e.g., an antibody or antigen-binding portion thereof, which are characterized by an acidic charge. For example, in monoclonal antibody (mAb) preparations, such acidic species can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing).

Acidic species of an antibody include charge variants, structure variants, and/or fragmentation variants. Exemplary charge variants include, but are not limited to, deamidation variants, afucosylation variants, methylglyoxal (MGO) variants, glycation variants, and citric acid variants. Exemplary structure variants include, but are not limited to, glycosylation variants and acetonation variants. Exemplary fragmentation variants include any truncated protein species from the target molecule due to dissociation of peptide chain, enzymatic and/or chemical modifications, including, but not limited to, Fc and Fab fragments, fragments missing a Fab, fragments missing a heavy chain variable domain, C-terminal truncation variants, variants with excision of N-terminal Asp in the light chain, and variants having N-terminal truncation of the light chain. Other acidic species variants include variants containing unpaired disulfides, host cell proteins, and host nucleic acids, chromatographic materials, and media components.

In certain embodiments, a protein composition can comprise more than one type of acidic species variant. For example, but not by way of limitation, the total acidic species can be divided based on chromatographic residence time. For example, as disclosed in FIG. 1, the total acidic species associated with the expression of adalimumab may be divided into a first acidic species region (AR1) and a second acidic species region (AR2). AR1 may comprise, for example, charge variants such as deamidation variants, MGO modified species, glycation variants, and citric acid variants, structural variants such as glycosylation variants and acetonation variants, and/or fragmentation variants. Other acidic variants such as host cells and unknown species may also be present. AR2 may comprise, for example, charge variants such as glycation variants and deamidation variants.

The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., TNFα) and still contain at least one heavy chain. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include a Fv fragment consisting of the VL and VH domains of a single arm of an antibody or a halfbody (as described in, for example, PCT Publication No. WO12/088,302, the entire contents of which are incorporated herein by reference). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York. 790 pp. (ISBN 3-540-41354-5).

The term “antibody construct” as used herein refers to a polypeptide comprising one or more of the antigen binding portions of the invention linked to a linker polypeptide or an immunoglobulin constant domain. Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions. Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences are known in the art.

An antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.

An “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds human TNFα). An isolated antibody that specifically binds TNFα may, however, have cross-reactivity to other antigens, such as the TNFα molecules from other species. Alternatively, an isolated antibody, or antigen-binding portion thereof, may not cross-react with the TNFα molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.

The term “human antibody”, as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell. In another embodiment, the human monoclonal antibodies are produced by phage display technologies as described, for example, in the Examples section below.

The term “recombinant human antibody”, as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library (Hoogenboom H. R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., (2002) Clin. Biochem. 35:425-445; Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29:128-145; Hoogenboom H., and Chames P. (2000) Immunology Today 21:371-378), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., U.S. Pat. No. 6,713,610; Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295; Kellermann S-A., and Green L. L. (2002) Current Opinion in Biotechnology 13:593-597; Little M. et al (2000) Immunology Today 21:364-370) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.

The term “human antibody derivatives” refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody.

The term “chimeric antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.

The term “CDR-grafted antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.

The term “humanized antibody” refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences. Such antibodies were generated by obtaining murine anti-TNFα monoclonal antibodies using traditional hybridoma technology followed by humanization using in vitro genetic engineering.

The term “antibody mimetic” or “antibody mimic” is intended to refer to molecules capable of mimicking an antibody's ability to bind an antigen, but which are not limited to native antibody structures. Examples of such antibody mimetics include, but are not limited to, Adnectins (i.e., fibronectin based binding molecules), Affibodies, DARPins, Anticalins, Avimers, and Versabodies all of which employ binding structures that, while they mimic traditional antibody binding, are generated from and function via distinct mechanisms. The embodiments of the instant invention, as they are directed to antibodies, or antigen binding portions thereof, also apply to the antibody mimetics described above.

As used herein, “isotype” refers to an antibody class (e.g., IgM or IgG1) that is encoded by the heavy chain constant region genes.

The terms “Kabat numbering”, “Kabat definitions and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.

As used herein, the terms “acceptor” and “acceptor antibody” refer to the antibody or nucleic acid sequence providing or encoding at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the amino acid sequences of one or more of the framework regions. In some embodiments, the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding the constant region(s). In yet another embodiment, the term “acceptor” refers to the antibody amino acid or nucleic acid sequence providing or encoding one or more of the framework regions and the constant region(s). In a specific embodiment, the term “acceptor” refers to a human antibody amino acid or nucleic acid sequence that provides or encodes at least 80%, or, at least 85%, at least 90%, at least 95%, at least 98%, or 100% of the amino acid sequences of one or more of the framework regions. In accordance with this embodiment, an acceptor may contain at least 1, at least 2, at least 3, least 4, at least 5, or at least 10 amino acid residues that does (do) not occur at one or more specific positions of a human antibody. An acceptor framework region and/or acceptor constant region(s) may be, e.g., derived or obtained from a germline antibody gene, a mature antibody gene, a functional antibody (e.g., antibodies well-known in the art, antibodies in development, or antibodies commercially available).

As used herein, the term “CDR” refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions. The term “CDR set” as used herein refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987) and (1991)) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia &Lesk, J. Mol. Biol. 196:901-917 (1987) and Chothia et al., Nature 342:877-883 (1989)) found that certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub-portions were designated as L1, L2 and L3 or H1, H2 and H3 where the “L” and the “H” designates the light chain and the heavy chains regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described by Padlan (FASEB J. 9:133-139 (1995)) and MacCallum (J Mol Biol 262(5):732-45 (1996)). Still other CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. The methods used herein may utilize CDRs defined according to any of these systems, although preferred embodiments use Kabat or Chothia defined CDRs.

As used herein, the term “canonical” residue refers to a residue in a CDR or framework that defines a particular canonical CDR structure as defined by Chothia et al. (J. Mol. Biol. 196:901-907 (1987); Chothia et al., J. Mol. Biol. 227:799 (1992), both are incorporated herein by reference). According to Chothia et al., critical portions of the CDRs of many antibodies have nearly identical peptide backbone confirmations despite great diversity at the level of amino acid sequence. Each canonical structure specifies primarily a set of peptide backbone torsion angles for a contiguous segment of amino acid residues forming a loop.

As used herein, the terms “donor” and “donor antibody” refer to an antibody providing one or more CDRs. In one embodiment, the donor antibody is an antibody from a species different from the antibody from which the framework regions are obtained or derived. In the context of a humanized antibody, the term “donor antibody” refers to a non-human antibody providing one or more CDRs.

As used herein, the term “framework” or “framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to correspondingly different interpretations. The six CDRs (CDR-L1, CDR-L2, and CDR-L3 of light chain and CDR-H1, CDR-H2, and CDR-H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the particular sub-regions as FR1, FR2, FR3 or FR4, a framework region, as referred by others, represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain. As used herein, a FR represents one of the four sub-regions, and FRs represents two or more of the four sub-regions constituting a framework region.

Human heavy chain and light chain acceptor sequences are known in the art.

As used herein, the term “germline antibody gene” or “gene fragment” refers to an immunoglobulin sequence encoded by non-lymphoid cells that have not undergone the maturation process that leads to genetic rearrangement and mutation for expression of a particular immunoglobulin. (See, e.g., Shapiro et al., Crit. Rev. Immunol. 22(3): 183-200 (2002); Marchalonis et al., Adv Exp Med. Biol. 484:13-30 (2001)). One of the advantages of germline antibody genes stems from the recognition that germline antibody genes are more likely than mature antibody genes to conserve essential amino acid sequence structures characteristic of individuals in the species, hence less likely to be recognized as from a foreign source when used therapeutically in that species.

As used herein, the term “key” residues refer to certain residues within the variable region that have more impact on the binding specificity and/or affinity of an antibody, in particular a humanized antibody. A key residue includes, but is not limited to, one or more of the following: a residue that is adjacent to a CDR, a potential glycosylation site (can be either N- or O-glycosylation site), a rare residue, a residue capable of interacting with the antigen, a residue capable of interacting with a CDR, a canonical residue, a contact residue between heavy chain variable region and light chain variable region, a residue within the Vernier zone, and a residue in the region that overlaps between the Chothia definition of a variable heavy chain CDR1 and the Kabat definition of the first heavy chain framework.

As used herein, the term “humanized antibody” is an antibody or a variant, derivative, analog or fragment thereof which binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody. As used herein, the term “substantially” in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to the amino acid sequence of a non-human antibody CDR. A humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab′, F(ab′)2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. A humanized antibody may also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In some embodiments, a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include the CH1, hinge, CH2, CH3, and CH4 regions of the heavy chain. In some embodiments, a humanized antibody only contains a humanized light chain. In some embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.

The humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including without limitation IgG 1, IgG2, IgG3 and IgG4. The humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well-known in the art.

The framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. Such mutations, however, will not be extensive. Usually, at least 80%, at least 85%, at least 90%, and at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences. As used herein, the term “consensus framework” refers to the framework region in the consensus immunoglobulin sequence. As used herein, the term “consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.

As used herein, “Vernier” zone refers to a subset of framework residues that may adjust CDR structure and fine-tune the fit to antigen as described by Foote and Winter (1992, J. Mol. Biol. 224:487-499, which is incorporated herein by reference). Vernier zone residues form a layer underlying the CDRs and may impact on the structure of CDRs and the affinity of the antibody.

The term “multivalent binding protein” is used in this specification to denote a binding protein comprising two or more antigen binding sites. The multivalent binding protein is engineered to have the three or more antigen binding sites, and is generally not a naturally occurring antibody. The term “multispecific binding protein” refers to a binding protein capable of binding two or more related or unrelated targets. Dual variable domain (DVD) binding proteins as used herein, are binding proteins that comprise two or more antigen binding sites and are tetravalent or multivalent binding proteins. Such DVDs may be monospecific, i.e. capable of binding one antigen or multispecific, i.e. capable of binding two or more antigens. DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to a DVD Ig. Each half of a DVD Ig comprises a heavy chain DVD polypeptide, and a light chain DVD polypeptide, and two antigen binding sites. Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.

As used herein, the term “neutralizing” refers to neutralization of biological activity of TNFα. A neutralizing binding protein is a neutralizing antibody whose binding to TNFα and/or a mutant TNFα protein results in inhibition of a biological activity of TNFα and/or the mutant TNFα. The neutralizing binding protein binds TNFα and/or a mutant TNFα protein and reduces a biologically activity of TNFα and/or a mutant TNFα protein by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% or more Inhibition of a biological activity of TNFα and/or a mutant TNFα protein by a neutralizing binding protein can be assessed by measuring one or more indicators of TNFα and/or mutant TNFα biological activity well known in the art.

The term “activity” includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-TNFα antibody that binds to a TNFα antigen and/or the neutralizing potency of an antibody, for example, an anti-TNFα antibody whose binding to TNFα inhibits the biological activity of TNFα.

The term “epitope” includes any polypeptide determinant capable of specific binding to an immunoglobulin or T-cell receptor. In certain embodiments, epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and/or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody. In certain embodiments, an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.

The term “surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jonsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277.

The terms “specific binding” or “specifically binding”, as used herein, in reference to the interaction of an antibody with another moiety, e.g., TNFα, mean an interaction that is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the moiety, e.g., TNFα. For example, an antibody recognizes and binds to a specific protein structure rather than to proteins, generally. If an antibody is specific for epitope “A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled “A” and the antibody, will reduce the amount of labeled A bound to the antibody.

As used herein, an antibody that “binds” or “specifically binds” to an antigen, e.g., TNFα, is intended to refer to an antibody, or antigen-binding portion thereof, that specifically binds to the antigen. The term “Kon” (also “Kon”, “kon”), as used herein, is intended to refer to the on rate constant for association of a binding protein of the invention (e.g., an antibody of the invention) to an antigen to form an association complex, e.g., antibody/antigen complex, as is known in the art. The “Kon” also is known by the terms “association rate constant”, or “ka”, as used interchangeably herein. This value indicates the binding rate of an antibody to its target antigen or the rate of complex formation between an antibody and antigen as is shown by the equation below:
Antibody (“Ab”)+Antigen (“Ag”)→Ab-Ag.

The term “Koff” (also “Koff”, “koff”), as used herein, is intended to refer to the off rate constant for dissociation, or “dissociation rate constant”, of a binding protein of the invention (e.g., an antibody of the invention) from an association complex (e.g., an antibody/antigen complex) as is known in the art. This value indicates the dissociation rate of an antibody from its target antigen or separation of Ab-Ag complex over time into free antibody and antigen as shown by the equation below:
Ab+Ag←Ab-Ag.

The term “KD” (also “Kd”), as used herein, is intended to refer to the “equilibrium dissociation constant”, and refers to the value obtained in a titration measurement at equilibrium, or by dividing the dissociation rate constant (Koff) by the association rate constant (Kon). The association rate constant (Kon), the dissociation rate constant (Koff), and the equilibrium dissociation constant (K are used to represent the binding affinity of an antibody to an antigen. Methods for determining association and dissociation rate constants are well known in the art. Using fluorescence-based techniques offers high sensitivity and the ability to examine samples in physiological buffers at equilibrium. Other experimental approaches and instruments such as a BIAcore® (biomolecular interaction analysis) assay can be used (e.g., instrument available from BIAcore International AB, a GE Healthcare company, Uppsala, Sweden). Additionally, a KinExA® (Kinetic Exclusion Assay) assay, available from Sapidyne Instruments (Boise, Id.) can also be used.

The term “labeled binding protein” as used herein, refers to a protein with a label incorporated that provides for the identification of the binding protein. In one aspect, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 35S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 166Ho, or 153Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates. Representative examples of labels commonly employed for immunoassays include moieties that produce light, e.g., acridinium compounds, and moieties that produce fluorescence, e.g., fluorescein. Other labels are described herein. In this regard, the moiety itself may not be detectably labeled but may become detectable upon reaction with yet another moiety. Use of the term “detectably labeled” is intended to encompass the latter type of detectable labeling.

The term “antibody conjugate” refers to a binding protein, such as an antibody, linked, e.g., chemically linked, to a second chemical moiety, such as a therapeutic or cytotoxic agent. The term “agent” is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials. In one aspect, the therapeutic or cytotoxic agents include, but are not limited to, pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.

The term “polynucleotide” as referred to herein, means a polymeric form of two or more nucleotides, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes single and double stranded forms of DNA.

The term “isolated polynucleotide” as used herein shall mean a polynucleotide (e.g., of genomic, cDNA, or synthetic origin, or some combination thereof) that, by virtue of its origin, is not associated with all or a portion of a polynucleotide with which the “isolated polynucleotide” is found in nature; is operably linked to a polynucleotide to which it is not linked in nature; or does not occur in nature as part of a larger sequence.

The term “vector”, as used herein, is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.

The term “operably linked” refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences. “Operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term “expression control sequence” as used herein refers to polynucleotide sequences which are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The term “control sequences” is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. Protein constructs of the present invention may be expressed, and purified using expression vectors and host cells known in the art, including expression cassettes, vectors, recombinant host cells and methods for the recombinant expression and proteolytic processing of recombinant polyproteins and pre-proteins from a single open reading frame (e.g., WO 2007/014162, the entire contents of which are incorporated herein by reference).

“Transformation”, as defined herein, refers to any process by which exogenous DNA enters a host cell. Transformation may occur under natural or artificial conditions using various methods well known in the art. Transformation may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method is selected based on the host cell being transformed and may include, but is not limited to, viral infection, electroporation, lipofection, and particle bombardment. Such “transformed” cells include stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome. They also include cells which transiently express the inserted DNA or RNA for limited periods of time.

The term “recombinant host cell” (or simply “host cell”), as used herein, is intended to refer to a cell into which exogenous DNA has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell, but, to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. Host cells may include prokaryotic and eukaryotic cells selected from any of the Kingdoms of life. In one aspect, eukaryotic cells include protist, fungal, plant and animal cells. In a particular aspect, host cells include but are not limited to the prokaryotic cell line E. Coli; mammalian cell lines CHO, HEK 293 and COS; the insect cell line Sf9; and the fungal cell Saccharomyces cerevisiae.

Standard techniques may be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.

The terms “regulate” and “modulate” as used interchangeably, and refer to a change or an alteration in the activity of a molecule of interest (e.g., the biological activity of TNFα). Modulation may be an increase or a decrease in the magnitude of a certain activity or function of the molecule of interest. Exemplary activities and functions of a molecule include, but are not limited to, binding characteristics, enzymatic activity, cell receptor activation, and signal transduction.

As used herein, the term “effective amount” refers to the amount of a therapy which is sufficient to reduce or ameliorate the severity and/or duration of a disorder or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g., prophylactic or therapeutic agent). In one embodiment, an “effective amount” refers to the amount of an antibody, or antigen-binding portion thereof, of the invention, e.g., an anti-TNFα antibody, or antigen-binding portion thereof, that is sufficient to treat a disorder in which TNFα activity is detrimental.

As used herein, the term “a disorder in which TNFα activity is detrimental” is intended to include diseases and other disorders in which the presence of TNFα in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which TNFα activity is detrimental is a disorder in which inhibition of TNFα activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of TNFα in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of TNFα in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-TNFα antibody. There are numerous examples of disorders in which TNFα activity is detrimental described in more detail below. In one embodiment, the disorder in which TNFα activity is detrimental is selected from the group consisting of an autoimmune disorder, an intestinal disorder, and a skin disease. In one embodiment, the autoimmune disorder is selected from the group consisting of rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis, an allergy, multiple sclerosis, psoriatic arthritis, autoimmune diabetes, autoimmune uveitis, nephrotic syndrome and juvenile rheumatoid arthritis. In one embodiment, the intestinal disorder is Crohn's disease. In one embodiment, the skin disease is psoriasis. The use of TNFα antibodies and antibody portions obtained using methods of the invention for the treatment of specific disorders is discussed in detail further below.

Various aspects of the invention are described in further detail in the following subsections:

I. Antibodies, and Antigen-Binding Portions Thereof

The present invention provides antibodies, and antigen-binding portions thereof, having mutations to the C-terminal residues of their heavy chains that result in a decrease in the C-terminal processing of the heavy chains, thereby leading to more efficacious and stable antibodies and antibody compositions. The present invention also provides compositions comprising antibodies, and antigen-binding portions thereof, which exhibit decreased levels of C-terminal processing of lysines on the heavy chains. In one embodiment, the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof, such as adalimumab, modified according to the methods of the invention.

Exemplary antibodies are provided herein. The features of such exemplary antibodies are set forth in the herein and in the Sequence Listing, Figures, Tables and Examples.

1. C-Terminal Lysines

The present invention is based, at least in part, on the discovery that the C-terminal lysines of antibodies in pharmaceutical compositions can be lost during both the purification process and storage of the final composition as well as in vivo, resulting in compositions comprising individual antibody species that can vary at their C-terminus as to whether a lysine residue is present. This heterogeneity may lead to decreased efficacy and stability of the drug product. The inventors of the instant application have surprisingly discovered that modifying an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-lysine (“PGK”) by inserting a proline between the glycine and lysine (proline-glycine-proline-lysine, or “PGPK”) (SEQ ID NO:9) prevents the C-terminal processing of the antibody heavy chain and leads to more efficacious and stable antibody compositions. Without intending to be limited by theory, it is believed that these modifications make the antibody resistant to processing by a C-terminal carboxypeptidase (e.g., carboxypeptidase B or carboxypeptidase U).

As used herein, the term “modify”, “modifying” or “modified,” is intended to refer to changing one or more amino acids in an antibody, or antigen-binding portion thereof. The change can be produced by adding, substituting or deleting an amino acid at one or more positions. The change can be produced using standard techniques known in the art and described in more detail herein, such as PCR mutagenesis and site-directed mutagenesis. In one embodiment, of the invention, the C-terminal three amino acids of the heavy chain sequences of an antibody, or antigen-binding portion thereof, are modified from the native sequence of proline-glycine-lysine (“PGK”) (SEQ ID NO:10) to include a proline between the glycine and lysine, resulting in a C-terminal sequence of proline-glycine-proline-lysine (“PGPK”) (SEQ ID NO:9). In another embodiment, the C-terminal three amino acids of the heavy chain sequence of adalimumab are modified from the native sequence of proline-glycine-lysine (“PGK”) (see, e.g., SEQ ID NO:14) to include a proline between the glycine and lysine, resulting in a C-terminal sequence of proline-glycine-proline-lysine (“PGPK”) (see, e.g., SEQ ID NO:15).

As used herein, the term “lysine variant species” refers to an antibody, or antigen-binding portion thereof, comprising heavy chains with either zero, one or two C-terminal lysines. For example, the “Lys 0” variant comprises an antibody, or antigen-binding portion thereof, with heavy chains that do not comprise a C-terminal lysine. The “Lys 1” variant comprises an antibody, or antigen-binding portion thereof, with one heavy chain that comprises a C-terminal lysine. The “Lys 2” variant comprises an antibody with both heavy chains comprising a C-terminal lysine. Lysine variants can be detected, for example, by weak cation exchange chromatography (WCX) of the expression product of a host cell expressing the antibody, or antigen-binding portion thereof. For example, but not by way of limitation, FIG. 2 depicts WCX analysis of adalimumab wherein the three lysine variants, as well as two acidic species, are resolved from each other.

A composition of the invention may comprise more than one lysine variant species of an antibody, or antigen-binding portion thereof. For example, in one embodiment, the composition may comprise a Lys 2 variant of an antibody, or antigen-binding portion thereof. The composition may comprise a Lys 1 variant of an antibody, or antigen-binding portion thereof. The composition may comprise a Lys 0 variant of an antibody, or antigen-binding portion thereof. In another embodiment, the composition may comprise both Lys 1 and Lys 2 variants, or Lys 0 and Lys 2 variants, or Lys 0 and Lys 1 variants of an antibody, or antigen-binding portion thereof. In another embodiment, the composition may comprise all three lysine variant species, i.e., Lys 0, Lys 1 and Lys 2, of an antibody, or antigen-binding portion thereof.

In one embodiment, the invention comprises a composition comprising an antibody, or antigen-binding portion thereof, wherein the composition comprises less than about 50% lysine variant species that lack a C-terminal lysine (Lys 0). In another embodiment, the composition comprises less than about 49%, 48%, 47%, 46%, 45%, 44%, 43%, 42%, 41%, 40%, 39%, 38%, 37%, 36%, 35%, 34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% lysine variant species that lack a C-terminal lysine (“Lys 0”). In another embodiment, the composition comprises about 50% to about 0%, about 40% to about 10%, about 30% to about 20%, about 40% to about 20%, or about 30% to about 15% lysine variant species that lack a C-terminal lysine (Lys 0). In one embodiment, the composition comprises 0% lysine variant species that lack a C-terminal lysine (Lys 0). As used herein, the percent lysine variant species in the composition refers to the weight of the specific lysine variant species in a sample in relation to the weight of the total lysine variant species sum (i.e., the sum of Lys 0, Lys 1 and Lys 2) contained in the sample or composition. For example, the percent lysine variant species can be calculated using weak cation exchange chromatography such as WCX-10, as described herein.

In another embodiment, the composition comprises less than about 25% lysine variant species that have one C-terminal lysine (Lys 1). In another embodiment, the composition comprises less than about 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11% 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% lysine variant species that have one C-terminal lysine (Lys 1). In another embodiment, the composition comprises about 25% to about 0%, about 20% to about 5%, about 15% to about 10%, about 20% to about 10%, about 15% to about 5%, or about 25% to about 5% lysine variant species that have one C-terminal lysine (Lys 1). In one embodiment, the composition comprises 0% lysine variant species that have one C-terminal Lysine (Lys 1).

In another embodiment, the composition comprises at least about 70% lysine variant species that have two C-terminal lysines (Lys 2). In another embodiment, the composition comprises at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% lysine variant species that have two C-terminal lysines (Lys 2). In one embodiment, the composition comprises about 70% to about 100%, about 70% to about 90%, about 70% to about 80%, about 80% to about 100%, about 85% to about 100%, about 90% to about 100%, about 95% to about 100%, about 80% to about 90%, about 85% to about 95%, about 75% to about 85%, or about 97% to about 100% lysine variant species that have two C-terminal lysines (Lys 2). In one embodiment, the composition comprises 100% lysine variant species that have two C-terminal lysines (Lys 2).

In one embodiment, the composition may comprise less than about 10% acidic species, wherein the acidic species comprise a first acidic species region (AR1) and a second acidic species region (AR2). In another embodiment, the composition may comprise less than about 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or 0% acidic species. In another embodiment, the composition may comprise about 3% acidic species. In another embodiment, the composition may comprise less than about 1% AR1. In another embodiment, the composition may comprise about 0% AR1. In another embodiment, the composition may comprise less than about 5%, 4%, 3%, 2%, 1% or 0% AR2. In another embodiment, the composition may comprise about 0% AR1 and about 3% AR2. In yet another embodiment, the composition may comprise about 1% AR1 and about 4% AR2. As used herein, the percent AR in the composition refers to the weight of the acidic species in a sample in relation to the weight of the total antibodies contained in the sample. For example, the percent AR can be calculated using weak cation exchange chromatography such as WCX-10, as described herein.

In one embodiment of the invention, the antibody, or antigen-binding portion thereof, containing a PGPK modification is an anti-TNFα antibody, or antigen-binding portion thereof. For example, the invention comprises an anti-TNFα antibody, or antigen-binding portion thereof, comprising the full-length heavy and light chain sequences of adalimumab, except that the C-terminal three amino acids of the heavy chain sequences are modified from the native IgG1 sequence of PGK to include a proline between the glycine and lysine to result in a C-terminal sequence of PGPK. In certain embodiments, the instant invention is directed to an anti-TNFα antibody, or antigen-binding portion thereof, comprising the full-length heavy and light chain sequences of adalimumab, but which comprise a PGPK C-terminal sequence and at least one additional sequence modification to the heavy or light chain sequences. In certain embodiments, the additional sequence modification, or modifications, can include conservative or non-conservative substitutions, insertions, and/or deletions.

As used herein, the term “conservative sequence modifications” is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function using the functional assays described herein.

As used herein, the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % homology=# of identical positions/total # of positions×100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.

The percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4:11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package, using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.

In other embodiments, the present invention is also directed to nucleic acid sequences, such as DNA sequences and/or RNA sequences, encoding the antibodies, and antigen-binding portions thereof, of the present invention. For example, but not by way of limitation, the nucleic acid sequences of the present invention would include nucleic acid sequences encoding the full length heavy or light chain amino acid sequences of adalimumab, except that the C-terminal three amino acids of the heavy chain sequences is modified from the native antibody sequence of PGK to include a proline between the glycine and lysine to result in a C-terminal sequence of PGPK. Thus, in certain embodiments the nucleic acid sequences of the present invention include the addition of a CCC, CCU/T, CCA, or CCG codon (where U is employed in the codon if the nucleic acid is RNA and T is employed if the codon is DNA) between the sequences coding for the C-terminal lysine and the glycine immediately preceding it. In certain embodiments, the nucleic acids of the instant invention will comprise a sequence encoding a heavy chain C-terminal sequence of PGPK and at least one additional sequence modification to the heavy or light chain sequences. In certain embodiments, the additional sequence modification, or modifications, can include conservative or non-conservative substitutions, insertions, and/or deletions.

2. Variable Regions

The antigen-binding portion of an antibody comprises one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., human TNFα, or a portion thereof). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, the light chain variable domain (VL) and the heavy chain variable domain (VH), are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for function in the same manner as are intact antibodies. Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.

The antibodies of the present invention comprise at least one antigen binding domain. In one embodiment, the antibodies of the invention are anti-TNFα antibodies, or antigen-binding portions thereof.

In certain embodiments, the antibodies are human or chimeric antibodies or antibody fragments. In certain embodiments, the antibodies are human antibodies or antibody fragments. In other embodiments, the antibodies are human or chimeric antibodies or antibody fragments that bind to human TNFα and inhibit TNFα activity.

In certain embodiments, the antibodies comprise a VH and/or VL domain that has a given percent identify to at least one of the VH and/or VL sequences disclosed herein. As used herein, the term “percent (%) sequence identity”, also including “homology” is defined as the percentage of amino acid residues or nucleotides in a candidate sequence that are identical with the amino acid residues or nucleotides in the reference sequences after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Optimal alignment of the sequences for comparison may be produced, besides manually, by means of the local homology algorithm of Smith and Waterman, 1981, Ads App. Math. 2, 482, by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. MoI. Biol. 48, 443, by means of the similarity search method of Pearson and Lipman, 1988, Proc. Natl Acad. Sci. USA 85, 2444, or by means of computer programs which use these algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N and TFASTA in Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, Wis.).

The antibodies of the invention containing a PGPK modification may comprise, or have, a heavy chain variable region (HCVR) amino acid sequence having at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more, identity to the amino acid sequence of SEQ ID NO:2.

The antibodies of the invention containing a PGPK modification may comprise a HCVR amino acid sequence in which 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residue substitutions have been made relative to SEQ ID NO:2. The substitutions may be conservative amino acid substitutions. These antibodies may have at least two or more (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10 or more) of the biological characteristics described herein.

The antibodies of the invention containing a PGPK modification may comprise, or have, a light chain variable region (LCVR) domain amino acid sequence having at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO:1.

The antibodies of the invention containing a PGPK modification may comprise, or have, a LCVR amino acid sequence in which 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residue substitutions have been made relative to SEQ ID NO:1. In certain embodiments, the substitutions are conservative amino acid substitutions. These antibodies have at least two or more (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10 or more) of the biological characteristics described herein.

In a specific embodiment, the antibodies, or antigen-binding portions thereof, of the invention containing a PGPK modification may comprise, or have, a HCVR amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO:2, and a LCVR amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity to SEQ ID NO:1. These antibodies may have at least two more (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10 or more) of the TNFα biological characteristics described herein.

In certain embodiments, the antibodies containing a PGPK modification may comprise, or have, a HCVR amino acid sequence in which 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residue substitutions have been made relative to SEQ ID NO:2, and a LCVR amino acid sequence in which 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid residue substitutions have been made relative to SEQ ID NO:1. These antibodies may have at least two more (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10 or more) of the TNFα biological characteristics described herein.

3. Complementarity Determining Regions (CDRs)

Although the variable domain (VH and VL) comprises the antigen-binding region, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in segments called Complementarity Determining Regions (CDRs), both in the light chain (VL or VK) and the heavy chain (VH) variable domains. The more highly conserved portions of the variable domains are called the framework regions (FR). The variable domains of native heavy and light chains each comprise four FR, largely adopting a β-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the β-sheet structure. The CDRs in each chain are held together in close proximity by the FR and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see, Kabat et al., Supra). The three CDRs of the heavy chain are designated CDR-H1, CDR-H2, and CDR-H3, and the three CDRs of the light chain are designated CDR-L1, CDR-L2, and CDR-L3. The Kabat numbering system is used herein. As such, CDR-H1 begins at approximately amino acid 31 (i.e., approximately 9 residues after the first cysteine residue), includes approximately 5-7 amino acids, and ends at the next tyrosine residue. CDR-H2 begins at the fifteenth residue after the end of CDR-H1, includes approximately 16-19 amino acids, and ends at the next arginine or lysine residue. CDR-H3 begins at approximately the thirty third amino acid residue after the end of CDR-H2; includes 3-25 amino acids; and ends at the sequence W-G-X-G, where X is any amino acid. CDR-L1 begins at approximately residue 24 (i.e., following a cysteine residue); includes approximately 10-17 residues; and ends at the next tyrosine residue. CDR-L2 begins at approximately the sixteenth residue after the end of CDR-L1 and includes approximately 7 residues. CDR-L3 begins at approximately the thirty third residue after the end of CDR-L2; includes approximately 7-11 residues and ends at the sequence F-G-X-G, where X is any amino acid. Note that CDRs vary considerably from antibody to antibody (and by definition will not exhibit homology with the Kabat consensus sequences).

The antibodies of the invention comprise at least one antigen binding domain that comprises at least one complementarity determining region (CDR1, CDR2 and CDR3). In one embodiment, the antibodies comprise a VH that comprises at least one VH CDR (e.g., CDR-H1, CDR-H2 or CDR-H3). In another embodiment, the antibodies comprise a VL that comprises at least one VL CDR (e.g., CDR-L1, CDR-L2 or CDR-L3).

In certain embodiments, the antibodies of the invention containing a PGPK modification may comprise a combination of any CDR-H1 sequence disclosed herein; any CDR-H2 sequence disclosed herein; any CDR-H3 sequence disclosed herein; any CDR-L1 sequence disclosed herein; any CDR-L2 sequence disclosed herein; and any CDR-L3 sequence disclosed herein. In one embodiment, the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof. In certain embodiments, the antibody is an antibody fragment. In certain embodiments, the antibody is a human, humanized or chimeric antibody.

In certain embodiments, the anti-TNFα antibodies, or antigen-binding portions thereof, of the invention containing a PGPK modification may comprise, or have,

(a) a VH CDR1 having an amino acid sequence comprising SEQ ID NO:8,

(b) a VH CDR2 having an amino acid sequence comprising SEQ ID NO:6 and/or

(c) a VH CDR3 having an amino acid sequence comprising SEQ ID NO:4.

The anti-TNFα antibodies, or antigen-binding portions thereof, of the invention containing a PGPK modification may comprise, or have,

(a) a VH CDR1 having an amino acid sequence comprising SEQ ID NO:7,

(b) a VH CDR2 having an amino acid sequence comprising SEQ ID NO:5 and/or

(c) a VH CDR3 having an amino acid sequence comprising SEQ ID NO:3.

a. CDR3s

It is well known in the art that VH CDR3 and VL CDR3 domains play an important role in the binding specificity/affinity of an antibody for an antigen (Xu and Davis, Immunity, 13: 37-45, 2000).

Accordingly, in one embodiment, the antibodies, or antigen-binding portions thereof, containing a PGPK modification may comprise or have a VH CDR3 having an amino acid sequence identical to or comprising 1, 2, or 3 amino acid residue substitutions relative to SEQ ID NO:4. In one embodiment, the antibody may be an anti-TNFα antibody, or antigen-binding portion thereof. These anti-TNFα antibodies, or antigen-binding portions thereof, may have at least two or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10 or more) of the biological characteristics described herein.

In another embodiment, the antibodies, or antigen-binding portions thereof, containing a PGPK modification may comprise or have a VL CDR3 having an amino acid sequence identical to or comprising 1, 2, or 3 amino acid residue substitutions relative to SEQ ID NO:3. In one embodiment, the antibody may be an anti-TNFα antibody, or antigen-binding portion thereof. These anti-TNFα antibodies, or antigen-binding portions thereof, may have at least two or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10 or more) of the biological characteristics described herein.

In another embodiment, the anti-TNFα antibody, or antigen-binding portion thereof, containing a PGPK modification may comprise or have:

(a) a VH CDR3 having an amino acid sequence identical to or comprising 1, 2, or 3 amino acid residue substitutions relative to SEQ ID NO:4, and

(b) a VL CDR3 having an amino acid sequence identical to or comprising 1, 2, or 3 amino acid residue substitutions relative to SEQ ID NO:3.

The invention contemplates antibodies, and antigen-binding antibody fragments thereof, having any combination of the foregoing VH and VL CDR1s. For example, antibodies containing a PGPK modification may comprise:

(a) a VH CDR3 having a sequence identical to SEQ ID NO:4,

(b) a VL CDR3 having an amino acid sequence comprising one amino acid substitution relative to SEQ ID NO:3. All other combinations are similarly contemplated.

The present invention encompasses antibodies comprising amino acids in a sequence that is substantially the same as an amino acid sequence described herein. Amino acid sequences that are substantially the same as the sequences described herein include sequences comprising conservative amino acid substitutions, as well as amino acid deletions and/or insertions. A conservative amino acid substitution refers to the replacement of a first amino acid by a second amino acid that has chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity/hydrophilicity) that are similar to those of the first amino acid. Conservative substitutions include replacement of one amino acid by another within the following groups: lysine (K), arginine (R) and histidine (H); aspartate (D) and glutamate (E); asparagine (N), glutamine (Q), serine (S), threonine (T), tyrosine (Y), K, R, H, D and E; alanine (A), valine (V), leucine (L), isoleucine (I), proline (P), phenylalanine (F), tryptophan (W), methionine (M), cysteine (C) and glycine (G); F, W and Y; C, S and T. Similarly contemplated is replacing a basic amino acid with another basic amino acid (e.g., replacement among Lys, Arg, His), replacing an acidic amino acid with another acidic amino acid (e.g., replacement among Asp and Glu), replacing a neutral amino acid with another neutral amino acid (e.g., replacement among Ala, Gly, Ser, Met, Thr, Leu, Ile, Asn, Gln, Phe, Cys, Pro, Trp, Tyr, Val).

The foregoing applies equally to anti-TNFα antibodies and antibody fragments of the invention. Antibodies and antibody fragments having any one or more of the foregoing functional and structural characteristics are contemplated.

4. Framework Regions

The variable domains of the heavy and light chains each comprise four framework regions (FR1, FR2, FR3, FR4), which are the more highly conserved portions of the variable domains. The four FRs of the heavy chain are designated FR-H1, FR-H2, FR-H3 and FR-H4, and the four FRs of the light chain are designated FR-L1, FR-L2, FR-L3 and FR-L4. The Kabat numbering system is used herein, See Table 1, Kabat et al., Supra. As such, FR-H1 begins at position 1 and ends at approximately amino acid 30, FR-H2 is approximately from amino acid 36 to 49, FR-H3 is approximately from amino acid 66 to 94 and FR-H4 is approximately amino acid 103 to 113. FR-L1 begins at amino acid 1 and ends at approximately amino acid 23, FR-L2 is approximately from amino acid 35 to 49, FR-L3 is approximately from amino acid 57 to 88 and FR-L4 is approximately from amino acid 98 to 107. In certain embodiments the framework regions may contain substitutions according to the Kabat numbering system, e.g., insertion at 106A in FR-L1. In addition to naturally occurring substitutions, one or more alterations (e.g., substitutions) of FR residues may also be introduced in an anti-TNFα antibody. In certain embodiments, these result in an improvement or optimization in the binding affinity of the antibody for TNFα, for example one or more of human, mouse, or cynomolgous TNFα. Examples of framework region residues to modify include those which non-covalently bind antigen directly (Amit et al., Science, 233:747-753 (1986)); interact with/effect the conformation of a CDR (Chothia et al., J. Mol. Biol., 196:901-917 (1987)); and/or participate in the VL-VH interface (U.S. Pat. No. 5,225,539).

In another embodiment the FR may comprise one or more amino acid changes for the purposes of “germlining” For example, the amino acid sequences of selected antibody heavy and light chains are compared to germline heavy and light chain amino acid sequences and where certain framework residues of the selected VL and/or VH chains differ from the germline configuration (e.g., as a result of somatic mutation of the immunoglobulin genes used to prepare the phage library), it may be desirable to “backmutate” the altered framework residues of the selected antibodies to the germline configuration (i.e., change the framework amino acid sequences of the selected antibodies so that they are the same as the germline framework amino acid sequences). Such “backmutation” (or “germlining”) of framework residues can be accomplished by standard molecular biology methods for introducing specific mutations (e.g., site-directed mutagenesis; PCR-mediated mutagenesis, and the like). In one embodiment, the variable light and/or heavy chain framework residues are backmutated. In another embodiment, the variable heavy chain of an antibody of the invention is backmutated. In another embodiment, the variable heavy chain of an antibody of the invention comprises at least one, at least two, at least three, at least four or more backmutations.

In certain embodiments, the VH of an anti-TNFα antibody of the invention may comprise a FR1, FR2, FR3 and/or FR4 that has an amino acid sequence identity with the corresponding framework regions (i.e., FR1 of antibody X as compared to FR1 of antibody Y) of any one or more of the VH chains of the anti-TNFα antibodies described herein and set forth in the Sequence Listing, that is from about 65% to about 100%. In one embodiment, the anti-TNFα antibodies comprise, or have, a VH FR amino acid sequence (e.g., FR1, FR2, FR3 and/or FR4) having at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity with the corresponding FR of the VH set forth in SEQ ID NO:2.

In certain embodiments, the anti-TNFα antibodies containing a PGPK modification may comprise a VH FR (e.g., FR1, FR2, FR3 and/or FR4) having an amino acid sequence identical to, or comprising 1, 2 or 3 amino acid substitutions relative to, the corresponding FR of the VH set forth in SEQ ID NO:2.

In certain embodiments, the VL of an anti-TNFα antibody containing a PGPK modification of the invention may comprise a FR1, FR2, FR3 and/or FR4 that has an amino acid sequence identity with the corresponding framework regions (i.e., FR1 of antibody X as compared to FR1 of antibody Y) of any one or more of the VH chains of the anti-TNFα antibodies described herein and set forth in the Sequence Listing, that is from about 65% to about 100%. In one embodiment, the anti-TNFα antibodies containing a PGPK modification may comprise a VL FR amino acid sequence (e.g., FR1, FR2, FR3 and/or FR4) having at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity with the corresponding FR of the VL chains set forth in SEQ ID NO:1.

5. Nucleotide Sequences Encoding Antibodies

In addition to the amino acid sequences described above, the present invention further provides nucleotide sequences corresponding to the amino acid sequences and encoding the antibodies of the invention. In one embodiment, the present invention provides polynucleotides comprising a nucleotide sequence encoding an anti-TNFα antibody containing a PGPK modification described herein or fragments thereof. These include, but are not limited to, nucleotide sequences that code for the above referenced amino acid sequences. Thus, the present invention provides polynucleotide sequences encoding VH and VL domain regions including CDRs and FRs of antibodies described herein as well as expression vectors for their efficient expression in cells (e.g., mammalian cells). Methods of making the anti-TNFα antibodies using polynucleotides are described below in more detail and are known in the art. The foregoing polynucleotides encode anti-TNFα antibodies containing a PGPK modification and having the structural and/or functional features described herein.

The present invention also encompasses polynucleotides that hybridize under stringent hybridization conditions, e.g., as defined herein, to polynucleotides that encode an antibody of the invention.

Stringent hybridization conditions include, but are not limited to, hybridization to filter-bound DNA in 6× sodium chloride/sodium citrate (SSC) at about 45° C. followed by one or more washes in 0.2×SSC/0.1% SDS at about 50-65° C., highly stringent conditions such as hybridization to filter-bound DNA in 6×SSC at about 45° C. followed by one or more washes in 0.1×SSC/0.2% SDS at about 65° C., or any other stringent hybridization conditions known to those skilled in the art (see, for example, Ausubel, F. M. et al., eds. 1989 Current Protocols in Molecular Biology, vol. 1, Green Publishing Associates, Inc. and John Wiley and Sons, Inc., NY at pages 6.3.1 to 6.3.6 and 2.10.3).

In certain embodiments, the polynucleotide sequences of the invention may also comprise a nucleotide sequence encoding an anti-TNFα antibody containing a PGPK modification VH which has at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to the nucleic acid sequence encoding SEQ ID NO:2.

In certain embodiments, the polynucleotide sequences of the invention may also comprise a nucleotide sequence encoding an anti-TNFα antibody containing a PGPK modification VL which has at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to the nucleotide sequence encoding SEQ ID NO:1.

The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.

A polynucleotide encoding an antibody may also be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, in one aspect polyA+RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3′ and 5′ ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art.

Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.

6. Biological Characteristics of the Anti-TNFα Antibodies

The anti-TNFα antibodies, or antigen-binding fragments thereof, of the invention comprising a PGPK modification are characterized by, or may have one or more of the biological characteristics described herein. As used herein, the term “biological characteristics” of an antibody refers to any one of the biochemical, binding and functional characteristics, which are used to select antibodies for therapeutic, research, and diagnostic uses as described, for example, in U.S. Pat. Nos. 6,090,382; 6,258,562; 6,509,015; 7,223,394; 7,541,031; 7,588,761; 7,863,426; 7,919,264; 8,197,813; 8,206,714; 8,216,583; 8,420,081; 8,092,998; 8,093,045; 8,187,836; 8,372,400; 8,034,906; 8,436,149; 8,231,876; 8,414,894; 8,372,401, PCT Publication No. WO2012/065072, the entire contents of each which are expressly incorporated herein by reference. Such characteristics are also described, for example, in “Highlights of Prescribing Information” for HUMIRA® (adalimumab) Injection (Revised January 2008); and below.

The biochemical characteristics of the antibodies of the invention include, but are not limited to, isoelectric point (pI) and melting temperature (Tm). The binding characteristics of the antibodies of the invention include, but are not limited to, binding specificity, dissociation constant (Kd), or its inverse, association constant (Ka), or its component kon or koff rates, epitope, ability to distinguish between various forms and/or preparations of TNFα (e.g., recombinant, native, acetylated) and ability to bind soluble and/or immobilized antigen. Methods for measuring the characteristics of the antibodies are well known in the art, some of which are detailed below and in the examples section.

a. Biochemical Characteristics

Antibodies like all polypeptides have an Isoelectric Point (pI), which is generally defined as the pH at which a polypeptide carries no net charge. It is known in the art that protein solubility is typically lowest when the pH of the solution is equal to the isoelectric point (pI) of the protein. As used herein the pI value is defined as the pI of the predominant charge form. The pI of a protein may be determined by a variety of methods including but not limited to, isoelectric focusing and various computer algorithms (see, e.g., Bjellqvist et al., 1993, Electrophoresis 14:1023). In addition, the thermal melting temperatures (Tm) of the Fab domain of an antibody, can be a good indicator of the thermal stability of an antibody and may further provide an indication of the shelf-life. A lower Tm indicates more aggregation/less stability, whereas a higher Tm indicates less aggregation/more stability. Thus, in certain embodiments antibodies having higher Tm are desirable. Tm of a protein domain (e.g., a Fab domain) can be measured using any standard method known in the art, for example, by differential scanning calorimetry (see, e.g., Vermeer et al., 2000, Biophys. J. 78:394-404; Vermeer et al., 2000, Biophys. J. 79: 2150-2154).

Accordingly, in certain embodiments the present invention includes anti-TNFα antibodies containing a PGPK modification that have certain desirable biochemical characteristics such as a particular isoelectric point (pI) or melting temperature (Tm).

More specifically, in one embodiment, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a pI ranging from 5.5 to 9.5, e.g., about 5.5 to about 6.0, or about 6.0 to about 6.5, or about 6.5 to about 7.0, or about 7.0 to about 7.5, or about 7.5 to about 8.0, or about 8.0 to about 8.5, or about 8.5 to about 9.0, or about 9.0 to about 9.5. In other specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a pI that ranges from 5.5-6.0, or 6.0 to 6.5, or 6.5 to 7.0, or 7.0-7.5, or 7.5-8.0, or 8.0-8.5, or 8.5-9.0, or 9.0-9.5. Even more specifically, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a pI of at least 5.5, or at least 6.0, or at least 6.3, or at least 6.5, or at least 6.7, or at least 6.9, or at least 7.1, or at least 7.3, or at least 7.5, or at least 7.7, or at least 7.9, or at least 8.1, or at least 8.3, or at least 8.5, or at least 8.7, or at least 8.9, or at least 9.1, or at least 9.3, or at least 9.5. In other specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a pI of at least about 5.5, or at least about 6.0, or at least about 6.3, or at least about 6.5, or at least about 6.7, or at least about 6.9, or at least about 7.1, or at least about 7.3, or at least about 7.5, or at least about 7.7, or at least about 7.9, or at least about 8.1, or at least about 8.3, or at least about 8.5, or at least about 8.7, or at least about 8.9, or at least about 9.1, or at least about 9.3, or at least about 9.5.

It is possible to optimize solubility by altering the number and location of ionizable residues in the antibody to adjust the pI. For example the pI of a polypeptide can be manipulated by making the appropriate amino acid substitutions (e.g., by substituting a charged amino acid such as a lysine, for an uncharged residue such as alanine). Without wishing to be bound by any particular theory, amino acid substitutions of an antibody that result in changes of the pI of the antibody may improve solubility and/or the stability of the antibody. One skilled in the art would understand which amino acid substitutions would be most appropriate for a particular antibody to achieve a desired pI. In one embodiment, a substitution is generated in an antibody of the invention to alter the pI. It is specifically contemplated that the substitution(s) of the Fc region that result in altered binding to FcγR (described supra) may also result in a change in the pI. In another embodiment, substitution(s) of the Fc region are specifically chosen to effect both the desired alteration in FcγR binding and any desired change in pI.

In one embodiment, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a Tm ranging from 65° C. to 120° C. In specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a Tm ranging from about 75° C. to about 120° C., or about 75° C. to about 85° C., or about 85° C. to about 95° C., or about 95° C. to about 105° C., or about 105° C. to about 115° C., or about 115° C. to about 120° C. In other specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a Tm ranging from 75° C. to 120° C., or 75° C. to 85° C., or 85° C. to 95° C., or 95° C. to 105° C., or 105° C. to 115° C., or 115° C. to 120° C. In still other specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a Tm of at least about 65° C., or at least about 70° C., or at least about 75° C., or at least about 80° C., or at least about 85° C., or at least about 90° C., or at least about 95° C., or at least about 100° C., or at least about 105° C., or at least about 110° C., or at least about 115° C., or at least about 120° C. In yet other specific embodiments, the anti-TNFα antibodies of the present invention containing a PGPK modification may have a Tm of at least 65° C., or at least 70° C., or at least 75° C., or at least 80° C., or at least 85° C., or at least 90° C., or at least 95° C., or at least 100° C., or at least 105° C., or at least 110° C., or at least 115° C., or at least 120° C.

b. Binding Characteristics

As described above, the anti-TNFα antibodies of the invention containing a PGPK modification bind at least one epitope or antigenic determinant of TNFα or fragment thereof either exclusively or preferentially with respect to other polypeptides. The term “epitope” is defined above, and includes a protein determinant capable of binding to an antibody. These protein determinants or epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents. The term “discontinuous epitope” as used herein, refers to a conformational epitope on a protein antigen which is formed from at least two separate regions in the primary sequence of the protein. In certain embodiments, the antibodies of the invention bind to human TNFα and antigenic fragments thereof. In certain embodiments, the anti-TNFα antibodies bind the same epitope as an antibody comprising the six CDRs of any of the antibodies listed in the Examples or Sequence Listing.

The antibodies of the invention may bind epitopes conserved across species. For example, antibodies of the invention may bind murine, non-human primate, rat, bovine, pig or other mammalian TNFα and antigenic fragments thereof. In one embodiment, the antibodies of the invention may bind to one or more TNFα orthologs and or isoforms. In a specific embodiment, antibodies of the invention may bind to TNFα and antigenic fragments thereof from one or more species, including, but not limited to, mouse, rat, monkey, primate, and human. In certain embodiments, the antibodies of the invention may bind an epitope within humans across TNFα homologs and/or isoforms and/or conformational variants and/or subtypes.

The interactions between antigens and antibodies are the same as for other non-covalent protein-protein interactions. In general, four types of binding interactions exist between antigens and antibodies: (i) hydrogen bonds, (ii) dispersion forces, (iii) electrostatic forces between Lewis acids and Lewis bases, and (iv) hydrophobic interactions. Hydrophobic interactions are a major driving force for the antibody-antigen interaction, and are based on repulsion of water by non-polar groups rather than attraction of molecules (Tanford, 1978). However, certain physical forces also contribute to antigen-antibody binding, for example, the fit or complimentary of epitope shapes with different antibody binding sites. Moreover, other materials and antigens may cross-react with an antibody, thereby competing for available free antibody.

Measurement of the affinity constant and specificity of binding between antigen and antibody is a pivotal element in determining the efficacy of therapeutic, diagnostic and research methods using the anti-TNFα antibodies. “Binding affinity” generally refers to the strength of the sum total of the noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, “binding affinity” refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the equilibrium dissociation constant (Kd), which is calculated as the ratio koff/kon. See, e.g., Chen, Y., et al., (1999) J. Mol. Biol 293:865-881. Affinity can be measured by common methods known in the art, including those described and exemplified herein, such as BIACORE™. Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer.

The anti-TNFα antibodies of the present invention containing a PGPK modification may have binding affinities for a TNFα epitope that include a dissociation constant (Kd) of less than 1×10−2M, 1×10−3M, 1×10−4M, 1×10−5M, 1×10−6M, 1×10−7M, 1×10−8M, 1×10−9M, 1×10−10M, 1×10−11M, 1×10−12M, 1×10−13M, 1×10−14M or less than 1×10−15M. In one embodiment, the anti-TNFα antibodies containing a PGPK modification may have a Kd of less than 10−7M, less than 5×10−8M, less than 10−8M, less than 5×10−9M, less than 10−9M, less than 5×10−10M, less than 10−10M, less than 5×10−11M, less than 10−11M, less than 5×10−12M, less than 10−12M, less than 5×10−13M, less than 10−13M, less than 5×10−14M, less than 10−14M, less than 5×10−15M, or less than 10−15 M. In certain embodiments, anti-TNFα antibodies containing a PGPK modification may have binding affinities for a TNFα epitope that include a dissociation constant (Kd) of between 1×10−6M and 1×10−10M, 1×10−6M and 1×10−11M, 1×10−6M and 1×10−12M, 1×10−6M and 1×10−13M, 1×10−6M and 1×10−15M, 1×10−6M and 1×10−15M, 1×10−7M and 1×10−10M, 1×10−7M and 1×10−11M, 1×10−7M and 1×10−12M, 1×10−7M and 1×10−13M, 1×10−7M and 1×10−14M, 1×10−7M and 1×10−15M, 1×10−8M and 1×10−10M, 1×10−8M and 1×10−11M, 1×10−8M and 1×10−12M, 1×10−8M and 1×10−13M, 1×10−8M and 1×10−14M, 1×10−8M and 1×10−15M, 1×10−9M and 1×10−11M, 1×10−9M and 1×10−11M, 1×10−9M and 1×10−12M, 1×10−9M and 1×10−13M, 1×10−9M and 1×10−14M and 1×10−9M and 1×10−15M. In certain embodiments, Kd is measured by BIACORE™. In certain embodiments, Kd is measured by cell binding.

In certain embodiments, the anti-TNFα antibodies containing a PGPK modification are high-affinity antibodies. As used herein, the term “high affinity”, when referring to an IgG type antibody, refers to an antibody having a KD of 10−8 M or less, alternatively 10−9 M or less and alternatively 10−10 M or less for an antigen. However, “high affinity” binding can vary for other antibody isotypes. For example, “high affinity” binding for an IgM isotype refers to an antibody having a KD of 10−7 M or less, 10−8 M or less, or 10−9 M or less for an antigen.

In certain embodiments, the anti-TNFα antibodies containing a PGPK modification may have an affinity between 5 pM and 200 pM for active human TNFα, as assessed by plasmon resonance. In certain embodiments, the affinity is approximately 5, 10, 15, 20, 25, 50, 60, 70, 75, 80, 90, 100, etc. pM. In certain embodiments, the affinity is between about 5 pM and 50 pM. In certain embodiments, the affinity is between about 5 pM and 100 pM.

In certain embodiments, the anti-TNFα antibodies containing a PGPK modification are described as having a binding affinity of a specific molarity or better. “Or better” when used herein refers to a stronger binding, represented by a smaller numerical Kd value. For example, for an antibody which has an affinity for an antigen of “0.6 nM or better”, the antibody's affinity for the antigen is <0.6 nM, i.e., 0.59 nM, 0.58 nM, 0.57 nM etc. or any value less than 0.6 nM.

In an alternative embodiment, the affinity of the anti-TNFα antibodies containing a PGPK modification is described in terms of the association constant (Ka), which is calculated as the ratio kon/koff. In this instance the anti-TNFα antibodies containing a PGPK modification may have binding affinities for a TNFα epitope that include an association constant (Ka) of at least 1×102M−1, 1×103M−1, 1×104M−1, 1×105M−1, 1×106M−1, 1×107M−1, 1×108M−1, 1×109M−1, 1×1010M−1 1×1011M−1 1×1012M−1, 1×1013M−1, 1×1014M−1 or at least, 1×1015M−1. In one embodiment, the anti-KIT antibodies have a Ka of at least 107 M−1, at least 5×107 M−1, at least 108 M−1, at least 5×108 M−1, at least 109 M−1, at least 5×109 M−1, at least 1010 M−1, at least 5×1010 M−1, at least 1011 M−1, at least 5×1011 M−1, at least 1012 M−1, at least 5×1012 M−1, at least 1013M−1, at least 5×1013 M−1, at least 1014 M−1, at least 5×1014 M−1, at least 1015 M−1, or at least 5×1015 M−1. In certain embodiments, anti-TNFα antibodies containing a PGPK modification may have binding affinities for a TNFα epitope that include an association constant (Ka) of between 1×102M−1 and 1×103M−1, 1×102M−1 and 1×104M−1, 1×102M−1 and 1×105M−1, 1×102M−1 and 1×106M−1, 1×103M−1 and 1×104M−1, 1×103M−1 and 1×105M−1, 1×103M−1 and 1×106M−1, 1×104M−1 and 1×105M−1, 1×104M−1 and 1×106M−1 and 1×105M−1 and 1×106M−1.

In certain embodiments the rate at which the anti-TNFα antibodies containing a PGPK modification may dissociate from a TNFα epitope may be more relevant than the value of the Kd or the Ka. In this instance the anti-TNFα antibodies of the invention containing a PGPK modification may bind to TNFα, or a fragment thereof, with a koff of less than 10−2 s−1, less than 10−3 s−1, less than 5×10−3 s−1, less than 10−4 s−1, less than 5×10−4 s−1, less than 10−5 s−1, less than 5×10−5 s−1, less than 10−6 s−1, less than 5×10−6 s−1, less than 10−7 s−1, less than 5×10−7 s−1, less than 10−8 s−1, less than 5×10−8 s−1, less than 10−9 s−1, less than 5×10−9 s−1, or less than 10−10 s−1. In certain other embodiments the rate at which the anti-TNFα antibodies containing a PGPK modification may associate with a TNFα epitope may be more relevant than the value of the Kd or the Ka. In this instance the anti-TNFα antibodies of the invention containing a PGPK modification may bind to TNFα, or a fragment thereof, with a kon rate of at least 105 M−1 s−1, at least 5×105M−1 s−1, at least 106M−1 s−1, at least 5×106M−1 s−1, at least 107 M−1 s−1, at least 5×107M−1 s−1, or at least 108M−1 s−1, or at least 109M−1 s−1.

Determination of binding affinity can be measured using the specific techniques described further in the Example section, and methods well known in the art. One such method includes measuring the disassociation constant “Kd” by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay that measures solution binding affinity of Fabs for antigen by equilibrating Fab with a minimal concentration of (125I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (Chen, et al., (1999) J. Mol. Biol 293:865-881). To establish conditions for the assay, microtiter plates (Dynex) are coated overnight with 5 μg/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (H 9.6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23° C.). In a non-adsorbant plate (Nunc #269620), 100 pM 26 pM [125I]-antigen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of an anti-VEGF antibody, Fab-12, in Presta et al., (1997) Cancer Res. 57:4593-4599). The Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., 65 hours) to insure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% Tween-20 in PBS. When the plates have dried, 150 μl/well of scintillant (MicroScint-20; Packard) is added, and the plates are counted on a Topcount gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding are chosen for use in competitive binding assays.

The Kd value may also be measured by using surface plasmon resonance assays using a BIACORE™-2000 or a BIACORE™-3000 (BIAcore, Inc., Piscataway, N.J.) at 25° C. with immobilized antigen CM5 chips at ˜10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIAcore Inc.) are activated with N-ethyl-N′-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen is diluted with 110 mM sodium acetate, pH 4.8, into 5 ug/ml (˜0.2 uM) before injection at a flow rate of 5 ul/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, IM ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% Tween 20 (PBST) at 25° C. at a flow rate of approximately 25 ul/min Association rates (kon) and dissociation rates (koff) are calculated using a simple one-to-one Langmuir binding model (BIACORE™ Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgram.

If the on-rate exceeds 106 M−1S−1 by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation=295 nm; emission=340 nm, 16 nm band-pass) at 25° C. of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000-series SLM-Aminco spectrophotometer (ThermoSpectronic) with a stir red cuvette. An “on-rate” or “rate of association” or “association rate” or “kon” according to this invention can also be determined with the same surface plasmon resonance technique described above using a BIACORE™-2000 or a BIACORE™-3000 (BIAcore, Inc., Piscataway, N.J.) as described above.

Methods and reagents suitable for determination of binding characteristics of an antibody of the present invention, or an altered/mutant derivative thereof (discussed below), are known in the art and/or are commercially available (see, for example, U.S. Pat. Nos. 6,849,425; 6,632,926; 6,294,391; 6,143,574). Moreover, equipment and software designed for such kinetic analyses are commercially available (e.g., BIACORE® A100, and BIACORE® 2000 instruments; Biacore International AB, Uppsala, Sweden).

In one embodiment, a binding assay may be performed either as a direct binding assay or as a competition-binding assay. Binding can be detected using standard ELISA or standard Flow Cytometry assays. In a direct binding assay, a candidate antibody is tested for binding to TNFα antigen. Competition-binding assays, on the other hand, assess the ability of a candidate antibody to compete with a known anti-TNFα antibody or other compound that binds TNFα. In general, any method that permits the detection and/or measuring of binding of an antibody with TNFα antigen may be used for detecting and measuring the binding characteristics of the antibodies of the invention. One of skill in the art will recognize these well known methods and for this reason are not provided in detail here.

c. Functional Characteristics

In certain embodiments, the antibodies of the invention containing a PGPK modification are anti-TNFα antibodies which modulate, e.g., inhibit, human TNFα activity. In certain embodiments, the anti-TNFα antibodies of the instant invention will retain one or more specific anti-TNFα antibody activity. In certain embodiments, the anti-TNFα antibodies of the instant invention will retain activities such as the binding specificity/affinity of an anti-TNFα antibody for its antigen, e.g., an anti-TNFα antibody that binds to a TNFα antigen and/or the neutralizing potency of an antibody, e.g., an anti-TNFα antibody whose binding to hTNFα inhibits the biological activity of hTNFα.

In certain embodiments, the compositions of the present invention include anti-TNFα antibodies that dissociate from hTNFα with a Kd of about 1×10−8 M or less and a Koff rate constant of 1×10−3 s−1 or less, both determined, for example, but not by way of limitation, by surface plasmon resonance. In specific non-limiting embodiments, the anti-TNFα antibodies of the instant invention competitively inhibit the binding of adalimumab to TNFα under physiological conditions.

In one embodiment, the antibodies of the invention exhibit a reduced antibody related toxicity as compared to previously described antibodies.

In another embodiment, the antibodies of the invention exhibit increased tissue (e.g., cartilage) penetration, increased TNFα affinity, reduced cartilage destruction, reduced bone erosion, reduced synovial proliferation, reduced cell infiltration, reduced chondrocyte death, reduced proteoglycan loss, increased protection against the development of arthritis scores when administered to an animal model of arthritis, and/or increased protection against the development of histopathology scores when administered to an animal model of arthritis as compared to previously described antibodies, e.g., antibodies not containing a PGPK modification.

In certain embodiments, the antibodies of the invention may bind epitopes conserved across species. In one embodiment, antibodies of the invention bind murine, non-human primate, rat, bovine, pig or other mammalian TNFα and antigenic fragments thereof. In one embodiment the antibodies of the invention may bind to one or more TNFα orthologs and or isoforms. In a specific embodiment, antibodies of the invention bind to TNFα and antigenic fragments thereof from one or more species, including, but not limited to, mouse, rat, monkey, primate, and human. In certain embodiments, the antibodies of the invention may bind an epitope within humans across TNFα homologs and/or isoforms and/or conformational variants and/or subtypes.

II. Expression and Production of Antibodies

The following section describes exemplary techniques for the production of the antibodies, and antigen-binding portions thereof, containing a PGPK modification of the invention. Such techniques are merely exemplary.

To express an antibody, or antigen-binding portion thereof, comprising a PGPK modification of the invention, DNA(s) encoding the antibody, or antigen-binding portion thereof, such as DNA(s) encoding partial or full-length light and heavy chains, may be inserted into one or more expression vector such that the genes are operatively linked to transcriptional and translational control sequences (see, e.g., U.S. Pat. Nos. 6,090,382; 6,258,562; 6,509,015; 7,223,394; 7,541,031; 7,588,761; 7,863,426; 7,919,264; 8,197,813; 8,206,714; 8,216,583; 8,420,081; 8,092,998; 8,093,045; 8,187,836; 8,372,400; 8,034,906; 8,436,149; 8,231,876; 8,414,894; 8,372,401, and PCT Publication No. WO2012/065072, the entire contents of each which are expressly incorporated herein by reference in their entireties). In this context, the term “operatively linked” is intended to mean that a gene encoding the antibody, or antigen-binding portion thereof, of interest is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. In certain embodiments, the antibody, or antigen-binding portion thereof comprising a PGPK modification will comprise multiple polypeptides, such as the heavy and light chains. Thus, in certain embodiments, genes encoding multiple polypeptides, such as antibody light chain genes and antibody heavy chain genes, can be inserted into a separate vector or, more typically, the genes are inserted into the same expression vector. Genes are inserted into expression vectors by standard methods (e.g., ligation of complementary restriction sites on the gene fragment and vector, or blunt end ligation if no restriction sites are present). Prior to insertion of the gene or genes, the expression vector may already carry additional polypeptide sequences, such as, but no limited to, antibody constant region sequences. For example, one approach to converting an antibody or antibody-related VH and VL sequences to full-length antibody genes is to insert them into expression vectors already encoding heavy chain constant and light chain constant regions, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector and the VL segment is operatively linked to the CL segment within the vector. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the protein from a host cell. The gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).

In addition to protein coding genes, a recombinant expression vector of the invention can carry one or more regulatory sequence that controls the expression of the protein coding genes in a host cell. The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the protein coding genes. Such regulatory sequences are described, e.g., in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990), the entire teaching of which is incorporated herein by reference. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma. For further description of viral regulatory elements, and sequences thereof, see, e.g., U.S. Pat. No. 5,168,062 by Stinski, U.S. Pat. No. 4,510,245 by Bell et al. and U.S. Pat. No. 4,968,615 by Schaffner et al., the entire contents of which are expressly incorporated herein by reference.

In addition to the protein coding genes and regulatory sequences, a recombinant expression vector of the invention may carry one or more additional sequences, such as a sequence that regulates replication of the vector in host cells (e.g., origins of replication) and/or a selectable marker gene. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al., the entire contents of which are incorporated herein by reference). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Suitable selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).

An antibody, or antibody portion, comprising a PGPK modification of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered. Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. Nos. 6,090,382; 6,258,562; 6,509,015; 7,223,394; 7,541,031; 7,588,761; 7,863,426; 7,919,264; 8,197,813; 8,206,714; 8,216,583; 8,420,081; 8,092,998; 8,093,045; 8,187,836; 8,372,400; 8,034,906; 8,436,149; 8,231,876; 8,414,894; 8,372,401, the entire contents of each which are expressly incorporated herein by reference in their entireties.

For expression of an antibody, or antigen-binding portion thereof, comprising a PGPK modification of the invention, for example, the expression vector(s) encoding the protein is (are) transfected into a host cell by standard techniques. The various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is theoretically possible to express the proteins of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, such as mammalian host cells, is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active protein. Prokaryotic expression of protein genes has been reported to be ineffective for production of high yields of active protein (Boss and Wood (1985) Immunology Today 6:12-13, the entire contents of which are incorporated herein by reference).

Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis 41P disclosed in DD 266,710 published Apr. 12, 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. One suitable E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.

In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces such as Schwanniomyces occidentalis; and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.

Suitable host cells for the expression of glycosylated proteins, for example, glycosylated antibodies, are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.

Mammalian cells may be used for expression and production of the recombinant protein of the present invention, however other eukaryotic cell types can also be employed in the context of the instant invention. See, e.g., Winnacker, From Genes to Clones, VCH Publishers, N.Y., N.Y. (1987). Suitable mammalian host cells for expressing recombinant proteins according to the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and ChasM, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NS0 myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding protein genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/−DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.

Host cells are transformed with the above-described expression or cloning vectors for protein production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.

The host cells used to produce a protein may be cultured in a variety of media. Commercially available media such as Ham's F10™ (Sigma), Minimal Essential Medium™ (MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium™ (DMEM), (Sigma) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102:255 (1980), U.S. Pat. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Pat. No. Re. 30,985 may be used as culture media for the host cells, the entire teachings of which are incorporated herein by reference. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

Host cells can also be used to produce portions of intact proteins, for example, antibodies, including Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to an antigen. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the target antibody, depending on the specificity of the antibody of the invention, by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.

In a suitable system for recombinant expression of a protein, for example, an antibody, or antigen-binding portion thereof, a recombinant expression vector encoding the protein, for example, both an antibody heavy chain and an antibody light chain, is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the protein gene(s) are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the gene(s). The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the protein, for example, the antibody heavy and light chains, and intact protein, for example, an antibody, is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the protein from the culture medium.

When using recombinant techniques, the protein, for example, antibodies or antigen binding fragments thereof, can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. In one aspect, if the protein is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells (e.g., resulting from homogenization), can be removed, e.g., by centrifugation or ultrafiltration. Where the protein is secreted into the medium, supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit.

Some antibodies can be secreted directly from the cell into the surrounding growth media; others are made intracellularly. For antibodies made intracellularly, the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration. Where the antibody is secreted, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit. Where the antibody is secreted into the medium, the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration. Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention, described below.

Antibody Conjugates

The antibodies, or antigen-binding portions thereof, of the invention comprising a PGPK modification may be conjugated or covalently attached to a substance using methods well known in the art. In one embodiment, the attached substance is a therapeutic agent, a detectable label (also referred to herein as a reporter molecule) or a solid support. Suitable substances for attachment to antibodies include, but are not limited to, an amino acid, a peptide, a protein, a polysaccharide, a nucleoside, a nucleotide, an oligonucleotide, a nucleic acid, a hapten, a drug, a hormone, a lipid, a lipid assembly, a synthetic polymer, a polymeric microparticle, a biological cell, a virus, a fluorophore, a chromophore, a dye, a toxin, a hapten, an enzyme, an antibody, an antibody fragment, a radioisotope, solid matrixes, semi-solid matrixes and combinations thereof. In a specific embodiment, the attached substance is a toxin. In another embodiment, the attached substance is an anti-cancer agent. Methods for conjugation or covalently attaching another substance to an antibody are well known in the art.

The antibodies of the invention may also be conjugated to a solid support. Antibodies may be conjugated to a solid support as part of the screening and/or purification and/or manufacturing process. Alternatively antibodies of the invention may be conjugated to a solid support as part of a diagnostic method or composition. A solid support suitable for use in the present invention is typically substantially insoluble in liquid phases. A large number of supports are available and are known to one of ordinary skill in the art. Thus, solid supports include solid and semi-solid matrixes, such as aerogels and hydrogels, resins, beads, biochips (including thin film coated biochips), microfluidic chip, a silicon chip, multi-well plates (also referred to as microtitre plates or microplates), membranes, conducting and nonconducting metals, glass (including microscope slides) and magnetic supports. More specific examples of solid supports include silica gels, polymeric membranes, particles, derivatized plastic films, glass beads, cotton, plastic beads, alumina gels, polysaccharides such as Sepharose, poly(acrylate), polystyrene, poly(acrylamide), polyol, agarose, agar, cellulose, dextran, starch, FICOLL, heparin, glycogen, amylopectin, mannan, inulin, nitrocellulose, diazocellulose, polyvinylchloride, polypropylene, polyethylene (including poly(ethylene glycol)), nylon, latex bead, magnetic bead, paramagnetic bead, superparamagnetic bead, starch and the like.

In some embodiments, the solid support may include a reactive functional group, including, but not limited to, hydroxyl, carboxyl, amino, thiol, aldehyde, halogen, nitro, cyano, amido, urea, carbonate, carbamate, isocyanate, sulfone, sulfonate, sulfonamide, sulfoxide, etc., for attaching the antibodies of the invention.

A suitable solid phase support can be selected on the basis of desired end use and suitability for various synthetic protocols. For example, where amide bond formation is desirable to attach the antibodies of the invention to the solid support, resins generally useful in peptide synthesis may be employed, such as polystyrene (e.g., PAM-resin obtained from Bachem Inc., Peninsula Laboratories, etc.), POLYHIPE™ resin (obtained from Aminotech, Canada), polyamide resin (obtained from Peninsula Laboratories), polystyrene resin grafted with polyethylene glycol (TENTAGEL™, Rapp Polymere, Tubingen, Germany), polydimethyl-acrylamide resin (available from Milligen/Biosearch, California), or PEGA beads (obtained from Polymer Laboratories).

The antibodies of the invention may also be conjugated to labels for purposes of diagnostics and other assays wherein the antibody and/or its associated ligand may be detected. A label conjugated to an antibody and used in the present methods and compositions described herein, is any chemical moiety, organic or inorganic, that exhibits an absorption maximum at wavelengths greater than 280 nm, and retains its spectral properties when covalently attached to an antibody. Labels include, without limitation, a chromophore, a fluorophore, a fluorescent protein, a phosphorescent dye, a tandem dye, a particle, a hapten, an enzyme and a radioisotope.

In certain embodiments, the antibodies of the invention are conjugated to a fluorophore. As such, fluorophores used to label antibodies of the invention include, without limitation; a pyrene (including any of the corresponding derivative compounds disclosed in U.S. Pat. No. 5,132,432), an anthracene, a naphthalene, an acridine, a stilbene, an indole or benzindole, an oxazole or benzoxazole, a thiazole or benzothiazole, a 4-amino-7-nitrobenz-2-oxa-1,3-diazole (NBD), a cyanine (including any corresponding compounds in U.S. Pat. Nos. 6,977,305 and 6,974,873), a carbocyanine (including any corresponding compounds in U.S. Ser. No. 09/557,275; U.S. Pat. Nos. 4,981,977; 5,268,486; 5,569,587; 5,569,766; 5,486,616; 5,627,027; 5,808,044; 5,877,310; 6,002,003; 6,004,536; 6,008,373; 6,043,025; 6,127,134; 6,130,094; 6,133,445; and publications WO 02/26891, WO 97/40104, WO 99/51702, WO 01/21624; EP 1 065 250 A1), a carbostyryl, a porphyrin, a salicylate, an anthranilate, an azulene, a perylene, a pyridine, a quinoline, a borapolyazaindacene (including any corresponding compounds disclosed in U.S. Pat. Nos. 4,774,339; 5,187,288; 5,248,782; 5,274,113; and 5,433,896), a xanthene (including any corresponding compounds disclosed in U.S. Pat. Nos. 6,162,931; 6,130,101; 6,229,055; 6,339,392; 5,451,343; 5,227,487; 5,442,045; 5,798,276; 5,846,737; 4,945,171; U.S. Ser. Nos. 09/129,015 and 09/922,333), an oxazine (including any corresponding compounds disclosed in U.S. Pat. No. 4,714,763) or a benzoxazine, a carbazine (including any corresponding compounds disclosed in U.S. Pat. No. 4,810,636), a phenalenone, a coumarin (including an corresponding compounds disclosed in U.S. Pat. Nos. 5,696,157; 5,459,276; 5,501,980 and 5,830,912), a benzofuran (including an corresponding compounds disclosed in U.S. Pat. Nos. 4,603,209 and 4,849,362) and benzphenalenone (including any corresponding compounds disclosed in U.S. Pat. No. 4,812,409) and derivatives thereof. As used herein, oxazines include resorufins (including any corresponding compounds disclosed in U.S. Pat. No. 5,242,805), aminooxazinones, diaminooxazines, and their benzo-substituted analogs.

In a specific embodiment, the fluorophores conjugated to the antibodies described herein include xanthene (rhodol, rhodamine, fluorescein and derivatives thereof) coumarin, cyanine, pyrene, oxazine and borapolyazaindacene. In other embodiments, such fluorophores are sulfonated xanthenes, fluorinated xanthenes, sulfonated coumarins, fluorinated coumarins and sulfonated cyanines. Also included are dyes sold under the tradenames, and generally known as, Alexa Fluor, DyLight, Cy Dyes, BODIPY, Oregon Green, Pacific Blue, IRDyes, FAM, FITC, and ROX.

The choice of the fluorophore attached to the antibody will determine the absorption and fluorescence emission properties of the conjugated antibody. Physical properties of a fluorophore label that can be used for antibody and antibody bound ligands include, but are not limited to, spectral characteristics (absorption, emission and stokes shift), fluorescence intensity, lifetime, polarization and photo-bleaching rate, or combination thereof. All of these physical properties can be used to distinguish one fluorophore from another, and thereby allow for multiplexed analysis. In certain embodiments, the fluorophore has an absorption maximum at wavelengths greater than 480 nm. In other embodiments, the fluorophore absorbs at or near 488 nm to 514 nm (particularly suitable for excitation by the output of the argon-ion laser excitation source) or near 546 nm (particularly suitable for excitation by a mercury arc lamp). In other embodiment a fluorophore can emit in the NIR (near infra red region) for tissue or whole organism applications. Other desirable properties of the fluorescent label may include cell permeability and low toxicity, for example if labeling of the antibody is to be performed in a cell or an organism (e.g., a living animal).

In certain embodiments, an enzyme is a label and is conjugated to an antibody of the invention. Enzymes are desirable labels because amplification of the detectable signal can be obtained resulting in increased assay sensitivity. The enzyme itself does not produce a detectable response but functions to break down a substrate when it is contacted by an appropriate substrate such that the converted substrate produces a fluorescent, colorimetric or luminescent signal. Enzymes amplify the detectable signal because one enzyme on a labeling reagent can result in multiple substrates being converted to a detectable signal. The enzyme substrate is selected to yield the measurable product, e.g., colorimetric, fluorescent or chemiluminescence. Such substrates are extensively used in the art and are well known by one skilled in the art.

In one embodiment, colorimetric or fluorogenic substrate and enzyme combination uses oxidoreductases such as horseradish peroxidase and a substrate such as 3,3′-diaminobenzidine (DAB) and 3-amino-9-ethylcarbazole (AEC), which yield a distinguishing color (brown and red, respectively). Other colorimetric oxidoreductase substrates that yield detectable products include, but are not limited to: 2,2-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), o-phenylenediamine (OPD), 3,3′,5,5′-tetramethylbenzidine (TMB), o-dianisidine, 5-aminosalicylic acid, 4-chloro-1-naphthol. Fluorogenic substrates include, but are not limited to, homovanillic acid or 4-hydroxy-3-methoxyphenylacetic acid, reduced phenoxazines and reduced benzothiazines, including AMPLEX® Red reagent and its variants (U.S. Pat. No. 4,384,042) and reduced dihydroxanthenes, including dihydrofluoresceins (U.S. Pat. No. 6,162,931) and dihydrorhodamines including dihydrorhodamine 123. Peroxidase substrates that are tyramides (U.S. Pat. Nos. 5,196,306; 5,583,001 and 5,731,158) represent a unique class of peroxidase substrates in that they can be intrinsically detectable before action of the enzyme but are “fixed in place” by the action of a peroxidase in the process described as tyramide signal amplification (TSA). These substrates are extensively utilized to label targets in samples that are cells, tissues or arrays for their subsequent detection by microscopy, flow cytometry, optical scanning and fluorometry.

In another embodiment, a colorimetric (and in some cases fluorogenic) substrate and enzyme combination uses a phosphatase enzyme such as an acid phosphatase, an alkaline phosphatase or a recombinant version of such a phosphatase in combination with a colorimetric substrate such as 5-bromo-6-chloro-3-indolyl phosphate (BCIP), 6-chloro-3-indolyl phosphate, 5-bromo-6-chloro-3-indolyl phosphate, p-nitrophenyl phosphate, or o-nitrophenyl phosphate or with a fluorogenic substrate such as 4-methylumbelliferyl phosphate, 6,8-difluoro-7-hydroxy-4-methylcoumarinyl phosphate (DiFMUP, U.S. Pat. No. 5,830,912) fluorescein diphosphate, 3-O-methylfluorescein phosphate, resorufin phosphate, 9H-(1,3-dichloro-9,9-dimethylacridin-2-one-7-yl) phosphate (DDAO phosphate), or ELF 97, ELF 39 or related phosphates (U.S. Pat. Nos. 5,316,906 and 5,443,986).

Glycosidases, in particular beta-galactosidase, beta-glucuronidase and beta-glucosidase, are additional suitable enzymes. Appropriate colorimetric substrates include, but are not limited to, 5-bromo-4-chloro-3-indolyl beta-D-galactopyranoside (X-gal) and similar indolyl galactosides, glucosides, and glucuronides, o-nitrophenyl beta-D-galactopyranoside (ONPG) and p-nitrophenyl beta-D-galactopyranoside. In one embodiment, fluorogenic substrates include resorufin beta-D-galactopyranoside, fluorescein digalactoside (FDG), fluorescein diglucuronide and their structural variants (U.S. Pat. Nos. 5,208,148; 5,242,805; 5,362,628; 5,576,424 and 5,773,236), 4-methylumbelliferyl beta-D-galactopyranoside, carboxyumbelliferyl beta-D-galactopyranoside and fluorinated coumarin beta-D-galactopyranosides (U.S. Pat. No. 5,830,912).

Additional enzymes include, but are not limited to, hydrolases such as cholinesterases and peptidases, oxidases such as glucose oxidase and cytochrome oxidases, and reductases for which suitable substrates are known.

Enzymes and their appropriate substrates that produce chemiluminescence are desirable for some assays. These include, but are not limited to, natural and recombinant forms of luciferases and aequorins. Chemiluminescence-producing substrates for phosphatases, glycosidases and oxidases such as those containing stable dioxetanes, luminol, isoluminol and acridinium esters are additionally useful.

In another embodiment, haptens such as biotin, are also utilized as labels. Biotin is useful because it can function in an enzyme system to further amplify the detectable signal, and it can function as a tag to be used in affinity chromatography for isolation purposes. For detection purposes, an enzyme conjugate that has affinity for biotin is used, such as avidin-HRP. Subsequently a peroxidase substrate is added to produce a detectable signal.

Haptens also include hormones, naturally occurring and synthetic drugs, pollutants, allergens, affector molecules, growth factors, chemokines, cytokines, lymphokines, amino acids, peptides, chemical intermediates, nucleotides and the like.

In certain embodiments, fluorescent proteins may be conjugated to the antibodies as a label. Examples of fluorescent proteins include green fluorescent protein (GFP) and the phycobiliproteins and the derivatives thereof. The fluorescent proteins, especially phycobiliprotein, are particularly useful for creating tandem dye labeled labeling reagents. These tandem dyes comprise a fluorescent protein and a fluorophore for the purposes of obtaining a larger stokes shift wherein the emission spectra is farther shifted from the wavelength of the fluorescent protein's absorption spectra. This is particularly advantageous for detecting a low quantity of a target in a sample wherein the emitted fluorescent light is maximally optimized, in other words little to none of the emitted light is reabsorbed by the fluorescent protein. For this to work, the fluorescent protein and fluorophore function as an energy transfer pair wherein the fluorescent protein emits at the wavelength that the fluorophore absorbs at and the fluorophore then emits at a wavelength farther from the fluorescent proteins than could have been obtained with only the fluorescent protein. A particularly useful combination is the phycobiliproteins disclosed in U.S. Pat. Nos. 4,520,110; 4,859,582; 5,055,556 and the sulforhodamine fluorophores disclosed in U.S. Pat. No. 5,798,276, or the sulfonated cyanine fluorophores disclosed in U.S. Pat. Nos. 6,977,305 and 6,974,873; or the sulfonated xanthene derivatives disclosed in U.S. Pat. No. 6,130,101 and those combinations disclosed in U.S. Pat. No. 4,542,104. Alternatively, the fluorophore functions as the energy donor and the fluorescent protein is the energy acceptor.

In certain embodiments, the label is a radioactive isotope. Examples of suitable radioactive materials include, but are not limited to, iodine (121I, 123I, 125I, 131I), carbon (14C), sulfur (35S), tritium (3H), indium (111In, 112In, 113mIn, 115mIn), technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum (99Mo), xenon (135Xe), fluorine (18F), 153SM, 177Lu, 159Gd, 149Pm, 140La, 175Yb, 166Ho, 90Y, 47Sc, 186Re, 188Re, 142Pr, 105Rh and 97 Ru.

III. Antibody Purification and Isolation

Once an antibody of the invention, e.g., an anti-TNFα antibody comprising a PGPK modification, has been expressed or produced, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigens Protein A or Protein G, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. In certain embodiments, the invention provides methods and compositions for producing a purified or partially purified (e.g., process-related impurity-reduced and/or product-related substance-reduced) protein preparation from a mixture comprising a protein of interest, e.g., an anti-TNFα antibody, or antigen-binding portion thereof, and at least one process-related impurity or product-related substance. In certain embodiments, the compositions of the present invention include, but are not limited to, process-related impurity-reduced and/or product-related substance-reduced compositions comprising a protein of interest. Such process-related impurity-reduced and/or product-related substance-reduced compositions address the need for improved product characteristics, including, but not limited to, product stability, product safety and product efficacy.

In certain embodiments, the purification process of the invention begins at the separation step when the antibody has been produced using production methods described above and/or by alternative production methods conventional in the art. Once a clarified solution or mixture comprising the protein of interest, e.g., an antibody, has been obtained, separation of the protein of interest from process-related impurities, such as the other proteins produced by the cell, as well as any product-related substances such as charge variants and/or size variants (aggregates and fragments), can be performed. In certain non-limiting embodiments, such separation is performed using Protein A affinity chromatography followed by a displacement chromatographic step. Such chromatographic steps may include, but are not limited to, ion exchange, mixed mode exchange, hydrophobic interaction chromatography, etc., as described herein and in U.S. Provisional Patent Application 61/893,068, entitled “Low Acidic Species Compositions and Methods for Producing and Using the Same,” filed on Oct. 18, 2013, the entire contents of which are expressly incorporated herein by reference. In certain embodiments, a combination of one or more different purification techniques described herein, including, but not limited to, ion exchange separation step(s) and/or hydrophobic interaction separation step(s) can also be employed. Such additional purification steps separate mixtures of proteins on the basis of their charge, degree of hydrophobicity, and/or size. In one aspect of the invention, such additional separation steps are performed using chromatography, including hydrophobic, anionic or cationic, or mixed mode interaction. Several different chromatography resins are available for each of these techniques, allowing accurate tailoring of the purification scheme to the particular protein involved. The essence of each of the separation methods is that proteins can either traverse at different rates through a column, achieving a physical separation that increases as they pass further down the column, or to adhere selectively to the separation medium, being then differentially eluted by different solvents, or by the presence of a displacer (in the context of displacement chromatography). In some cases, the antibody is separated from impurities when the impurities specifically adhere to the column and the antibody does not, i.e., the antibody is present in the flow-through, while in other cases the antibody will adhere to the column, while the impurities flow-through.

1. Primary Recovery

In certain embodiments, the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix. In certain embodiments, the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris. Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000×g to approximately 12,750×g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.

In certain embodiments, the primary recovery will include the use of one or more depth filtration steps to clarify the sample matrix and thereby aid in purifying the antibodies of interest in the present invention. In other embodiments, the primary recovery will include the use of one or more depth filtration steps post centrifugation to further clarify the sample matrix. Non-limiting examples of depth filters that can be used in the context of the instant invention include the Millistak+X0HC, F0HC, D0HC, A1HC, B1HC depth filters (EMD Millipore), Cuno™ model 30/60ZA, 60/90 ZA, VR05, VR07, delipid depth filters (3M Corp.). A 0.2 μm filter such as Sartorius's 0.45/0.2 μm Sartopore™ bi-layer or Millipore's Express SHR or SHC filter cartridges typically follows the depth filters.

In certain embodiments, the primary recovery process can also be a point at which to reduce or inactivate viruses that can be present in the sample matrix. For example, any one or more of a variety of methods of viral reduction/inactivation can be used during the primary recovery phase of purification including heat inactivation (pasteurization), pH inactivation, solvent/detergent treatment, UV and γ-ray irradiation and the addition of certain chemical inactivating agents such as β-propiolactone or e.g., copper phenanthroline as in U.S. Pat. No. 4,534,972. In certain embodiments of the present invention, the sample matrix is exposed to detergent viral inactivation during the primary recovery phase. In other embodiments, the sample matrix may be exposed to low pH inactivation during the primary recovery phase.

In those embodiments where viral reduction/inactivation is employed, the sample mixture can be adjusted, as needed, for further purification steps. For example, following low pH viral inactivation, the pH of the sample mixture is typically adjusted to a more neutral pH, e.g., from about 4.5 to about 8.5, prior to continuing the purification process. Additionally, the mixture may be diluted with water for injection (WFI) to obtain a desired conductivity.

2. Protein A Affinity Chromatography

In certain embodiments, the primary recovery sample is subjected to Protein A affinity chromatography to substantially purify the antibody of interest away from host cell proteins (“HCPs”). There are a variety of commercial sources for Protein A resin. Suitable resins include, but not limited to, MabSelect SuRe™, MabSelect SuRe LX, MabSelect, MabSelect Xtra, rProtein A Sepharose from GE Healthcare, ProSep HC, ProSep Ultra, and ProSep Ultra Plus from EMD Millipore, MapCapture from Life Technologies.

In certain embodiments, the Protein A column can be equilibrated with a suitable buffer prior to sample loading. Following the loading of the column, the column can be washed one or multiple times using a suitable sets of buffers. The Protein A column can then be eluted using an appropriate elution buffer. The eluate can be monitored using techniques well known to those skilled in the art. The eluate fractions of interest can be collected and then prepared for further processing.

The Protein A eluate may subject to a viral inactivation step either by detergent or low pH, provided this step is not performed prior to the Protein A capture operation. A proper detergent concentration or pH and time can be selected to obtain desired viral inactivation results. After viral inactivation, the Protein A eluate is usually pH and/or conductivity adjusted for subsequent purification steps.

The Protein A eluate may be subjected to filtration through a depth filter to remove turbidity and/or various impurities from the antibody of interest prior to additional chromatographic polishing steps. Examples of depth filters include, but not limited to, Millistak+X0HC, F0HC, D0HC, A1HC, and B1HC Pod filters (EMD Millipore), or Zeta Plus 30ZA/60ZA, 60ZA/90ZA, delipid, VR07, and VR05 filters (3M). The Protein A eluate pool may need to be conditioned to proper pH and conductivity to obtain desired impurity removal and product recovery from the depth filtration step.

3. Ion Exchange Chromatography

In certain embodiments, an ion exchange step will occur after the above-described Protein A affinity and/or displacement chromatography steps, such that an eluate comprising the protein of interest is obtained. Ion exchange separation includes any method by which two substances are separated based on the difference in their respective ionic charges, and can employ either cationic exchange material or anionic exchange material.

The use of a cationic exchange material versus an anionic exchange material can be based on the local charges of the protein at a given solution condition. Therefore, it is within the scope of this invention to employ an anionic exchange step prior to or subsequent to the use of a displacement chromatography step, or a cationic exchange step prior to or subsequent to the use of a displacement chromatography step.

In performing the separation, the initial protein mixture can be contacted with the ion exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique.

For example, in the context of batch purification, ion exchange material is prepared in, or equilibrated to, the desired starting buffer. Upon preparation, or equilibration, a slurry of the ion exchange material is obtained. The protein of interest, e.g., an antibody, solution is contacted with the slurry to adsorb the protein of interest to be separated to the ion exchange material. The solution comprising the process-related impurities and product-related substances that do not bind to the ion exchange material is separated from the slurry, e.g., by allowing the slurry to settle and removing the supernatant. The slurry can be subjected to one or more wash steps. If desired, the slurry can be contacted with a solution of higher conductivity to desorb process-related impurities and product-related substances that have bound to the ion exchange material. In order to elute bound polypeptides, the salt concentration of the buffer can be increased.

In the context of chromatographic separation, a chromatographic apparatus, commonly cylindrical in shape, is employed to contain the chromatographic support material (e.g., ion exchange material) prepared in an appropriate buffer solution. The chromatographic apparatus, if cylindrical, can have a diameter of about 5 mm to about 50 mm, and a height of 5 cm to 1 m, and in certain embodiments, particularly for large scale processing, a height of <30 cm is employed. Once the chromatographic material is added to the chromatographic apparatus, a sample containing the protein of interest, e.g., an antibody, is contacted to the chromatographic material to adsorb the protein of interest to be separated to the chromatographic material. The solution comprising the process-related impurities and product-related substances that do not bind to the chromatographic material is separated from the material by washing the materials and collecting fractions from the bottom of the column. The chromatographic material can be subjected to one or more wash steps. If desired, the chromatographic material can be contacted with a solution of higher conductivity to desorb process-related impurities and product-related substances that have bound to the chromatographic material. In order to elute bound polypeptides, the salt concentration of the buffer can be increased.

Ion exchange chromatography separates molecules based on differences between the local charges of the proteins of interest and the local charges of the chromatographic material. A packed ion-exchange chromatography column or an ion-exchange membrane device can be operated either in bind-elute mode or flow-through mode. In the bind-elute mode, the column or the membrane device is first conditioned with a buffer with low ionic strength and proper pH under which the protein carries sufficient local opposite charge to the local charge of the material immobilized on the resin based matrix. During the feed load, the protein of interest will be adsorbed to the resin due to electrostatic attraction. After washing the column or the membrane device with the equilibration buffer or another buffer with different pH and/or conductivity, the product recovery is achieved by increasing the ionic strength (i.e., conductivity) of the elution buffer to compete with the solute for the charged sites of the ion exchange matrix. Changing the pH and thereby altering the charge of the solute is another way to achieve elution of the solute. The change in conductivity or pH may be gradual (gradient elution) or stepwise (step elution). In the flow-through mode, the column or the membrane device is operated at selected pH and conductivity such that the protein of interest does not bind to the resin or the membrane while the process-related impurities and/or product-related substances will be retained to the column or the membrane. The column is then regenerated before next use.

Anionic or cationic substituents may be attached to matrices in order to form anionic or cationic supports for chromatography. Non-limiting examples of anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (O) groups. Cationic substitutents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S). Cellulose ion exchange resins such as DE23™, DE32™, DE52™, CM-23™, CM-32™, and CM-52™ are available from Whatman Ltd. Maidstone, Kent, U.K. SEPHADEX®-based and -locross-linked ion exchangers are also known. For example, DEAE-, QAE-, CM-, and SP—SEPHADEX® and DEAE-, Q-, CM- and S-SEPHAROSE® and SEPHAROSE® Fast Flow, and Capto™ S are all available from GE Healthcare. Further, both DEAE and CM derivitized ethylene glycol-methacrylate copolymer such as TOYOPEARL™ DEAE-6505 or M and TOYOPEARL™ CM-650S or M are available from Toso Haas Co., Philadelphia, Pa., or Nuvia S and UNOSphere™ S from BioRad, Hercules, Calif., Eshmuno® S from EMD Millipore, Billerica, Calif.

This ion exchange step facilitates the purification of the antibody of interest by reducing impurities such as HCPs, DNA and aggregates. In certain aspects, the ion exchange column is an anion exchange column. For example, but not by way of limitation, a suitable resin for such an anion exchange column is Capto™ Q, Nuvia™ Q, Q Sepharose Fast Flow, and Poros HQ 50. These resins are available from commercial sources such as GE Healthcare, BioRad, or Life Technologies. This anion exchange chromatography process can be carried out at or around room temperature.

4. Mixed Mode Chromatography

Mixed mode chromatography, also referred to herein as “multimodal chromatography”, is a chromatographic strategy that utilizes a support comprising a ligand that is capable of providing at least two different, in certain embodiments co-operative, sites that interact with the substance to be bound. In certain embodiments, one of these sites gives an attractive type of charge-charge interaction between the ligand and the substance of interest and the other site provides for electron acceptor-donor interaction and/or hydrophobic and/or hydrophilic interactions. Electron donor-acceptor interactions include interactions such as hydrogen-bonding, π-π, cation-π, charge transfer, dipole-dipole, induced dipole etc. Mixed mode chromatographic supports include, but are not limited to, Nuvia C Prime, Toyo Pearl MX Trp 650M, and Eshmuno® HCX. Mixed mode chromatography can be combined with one or more other steps described herein, including, but not limited to, Protein A chromatography, ion exchange, mixed mode chromatography, hydrophobic interaction chromatography, etc.

In certain embodiments, the mixed mode chromatography resin is comprised of ligands coupled to an organic or inorganic support, sometimes denoted a base matrix, directly or via a spacer. The support may be in the form of particles, such as essentially spherical particles, a monolith, filter, membrane, surface, capillaries, etc. In certain embodiments, the support is prepared from a native polymer, such as cross-linked carbohydrate material, such as agarose, agar, cellulose, dextran, chitosan, konjac, carrageenan, gellan, alginate etc. To obtain high adsorption capacities, the support can be porous, and ligands are then coupled to the external surfaces as well as to the pore surfaces. Such native polymer supports can be prepared according to standard methods, such as inverse suspension gelation (S Hjerten: Biochim Biophys Acta 79(2), 393-398 (1964). Alternatively, the support can be prepared from a synthetic polymer, such as cross-linked synthetic polymers, e.g., styrene or styrene derivatives, divinylbenzene, acrylamides, acrylate esters, methacrylate esters, vinyl esters, vinyl amides etc. Such synthetic polymers can be produced according to standard methods, see e.g., “Styrene based polymer supports developed by suspension polymerization” (R Arshady: Chimica e L′Industria 70(9), 70-75 (1988)). Porous native or synthetic polymer supports are also available from commercial sources, such as Amersham Biosciences, Uppsala, Sweden.

5. Hydrophobic Interaction Chromatography

The antibodies of the invention containing a PGPK modification, e.g., anti-TNFα antibody, or antigen-binding portion thereof, may be purified by using a hydrophobic interaction chromatography (HIC) step in addition to a displacement chromatography step.

In performing the separation, the sample mixture is contacted with the HIC material, e.g., using a batch purification technique or using a column or membrane chromatography. Prior to HIC purification it may be desirable to adjust the concentration of the kosmotropic salt to achieve desired protein binding to the resin or the membrane.

Whereas ion exchange chromatography relies on the local charge of the protein of interest for selective separation, hydrophobic interaction chromatography employs the hydrophobic properties of the proteins to achieve selective separation. Hydrophobic groups on the protein interact with hydrophobic groups of the resin or the membrane. The more hydrophobic a protein is the stronger it will interact with the column or the membrane. Thus the HIC step removes process-related impurities (e.g., HCPs) as well as product-related substances (e.g., aggregates and fragments).

Like ion exchange chromatography, a HIC column or membrane device can also be operated in product a bind-elute mode, a flow-through, or a hybrid mode wherein the product exhibits reversible binding to the chromatographic material. The bind-elute mode of operation has been explained above. For flow-through, the protein sample typically contains a relatively low level of salt than that used in the bind-elute mode. During this loading process, process-related impurities and product-related substances will bind to the resin while product flows through the column. After loading, the column is regenerated with water and cleaned with caustic solution to remove the bound impurities before next use. When used in connection with a hybrid mode, the product can be immobilized on the chromatographic support in the presence of a loading buffer, but can be removed by successive washes of buffer identical to or substantially similar to the loading buffer. During this process, process-related impurities and product-relates substances will either bind to the chromatographic material or flow through with a profile distinct from the protein of interest.

As hydrophobic interactions are strongest at high ionic strength, this form of separation is conveniently performed following salt elution step, such as those that are typically used in connection with ion exchange chromatography. Alternatively, salts can be added into a low salt level feed stream before this step. Adsorption of the antibody to a HIC column is favored by high salt concentrations, but the actual concentrations can vary over a wide range depending on the nature of the protein of interest, salt type and the particular HIC ligand chosen. Various ions can be arranged in a so-called soluphobic series depending on whether they promote hydrophobic interactions (salting-out effects) or disrupt the structure of water (chaotropic effect) and lead to the weakening of the hydrophobic interaction. Cations are ranked in terms of increasing salting out effect as Ba2+; Ca2+; Mg2+; Li+; Cs+; Na+; K+; Rb+; NH4+, while anions may be ranked in terms of increasing chaotropic effect as PO43−; SO42−; CH3CO3; Cl; Br; NO3; ClO4; I; SCN.

In general, Na+, K+ or NH4+ sulfates effectively promote ligand-protein interaction in HIC. Salts may be formulated that influence the strength of the interaction as given by the following relationship: (NH4)2SO4>Na2SO4>NaCl>NH4Cl>NaBr>NaSCN. In general, salt concentrations of between about 0.75 M and about 2 M ammonium sulfate or between about 1 and 4 M NaCl are useful.

HIC media normally comprise a base matrix (e.g., cross-linked agarose or synthetic copolymer material) to which hydrophobic ligands (e.g., alkyl or aryl groups) are coupled. A suitable HIC media comprises an agarose resin or a membrane functionalized with phenyl groups (e.g., a Phenyl Sepharose™ from GE Healthcare or a Phenyl Membrane from Sartorius). Many HIC resins are available commercially. Examples include, but are not limited to, Capto Phenyl, Phenyl Sepharose™ 6 Fast Flow with low or high substitution, Phenyl Sepharose™ High Performance, Octyl Sepharose™ High Performance (GE Healthcare); Fractogel™ EMD Propyl or Fractogel™ EMD Phenyl (E. Merck, Germany); Macro-Prep™ Methyl or Macro-Prep™ t-Butyl columns (Bio-Rad, California); WP HI-Propyl (C3)™ (J. T. Baker, New Jersey); and Toyopearl™ ether, phenyl or butyl (TosoHaas, Pa.).

6. Viral Filtration

Viral filtration is a dedicated viral reduction step in the entire purification process. This step is usually performed post chromatographic polishing steps. Viral reduction can be achieved via the use of suitable filters including, but not limited to, Planova 20N™, 50 N or BioEx from Asahi Kasei Pharma, Viresolve™ filters from EMD Millipore, ViroSart CPV from Sartorius, or Ultipor DV20 or DV50™ filter from Pall Corporation. It will be apparent to one of ordinary skill in the art to select a suitable filter to obtain desired filtration performance.

7. Ultrafiltration/Diafiltration

Certain embodiments of the present invention employ ultrafiltration and diafiltration steps to further concentrate and formulate the protein of interest, e.g., an antibody of the invention. Ultrafiltration is described in detail in: Microfiltration and Ultrafiltration: Principles and Applications, L. Zeman and A. Zydney (Marcel Dekker, Inc., New York, N.Y., 1996); and in: Ultrafiltration Handbook, Munir Cheryan (Technomic Publishing, 1986; ISBN No. 87762-456-9). One filtration process is Tangential Flow Filtration as described in the Millipore catalogue entitled “Pharmaceutical Process Filtration Catalogue” pp. 177-202 (Bedford, Mass., 1995/96). In contrast, diafiltration is a method of using membrane filters to remove and exchange salts, sugars, and non-aqueous solvents, to separate free from bound species, to remove low molecular-weight species, and/or to cause the rapid change of ionic and/or pH environments. Examples of membrane cassettes suitable for the present invention include, but not limited to, Pellicon 2 or Pellicon 3 cassetts with 10 kD, 30 kD or 50 kD membranes from EMD Millipore, Kvick 10 kD, 30 kD or 50 kD membrane cassettes from GE Healthcare, and Centramate or Centrasette 10 kD, 30 kD or 50 kD cassettes from Pall Corporation.

8. Methods of Assaying Sample Purity

a. Assaying Host Cell Protein

The antibodies of the present invention containing a PGPK modification may be assayed for purity using methods known in the art and described herein. For example, residual levels of host cell protein (HCP) concentration in the isolated/purified antibody composition may be measured. As described above, HCPs are desirably excluded from the final target substance product. Exemplary HCPs include proteins originating from the source of the antibody production. Failure to identify and sufficiently remove HCPs from the target antibody may lead to reduced efficacy and/or adverse subject reactions.

As used herein, the term “HCP ELISA” refers to an ELISA where the second antibody used in the assay is specific to the HCPs produced from cells, e.g., CHO cells, used to generate the antibody of interest. The second antibody may be produced according to conventional methods known to those of skill in the art. For example, the second antibody may be produced using HCPs obtained by sham production and purification runs, i.e., the same cell line used to produce the antibody of interest is used, but the cell line is not transfected with antibody DNA. In an exemplary embodiment, the second antibody is produced using HCPs similar to those expressed in the cell expression system of choice, i.e., the cell expression system used to produce the target antibody.

Generally, HCP ELISA comprises sandwiching a liquid sample comprising HCPs between two layers of antibodies, i.e., a first antibody and a second antibody. The sample is incubated during which time the HCPs in the sample are captured by the first antibody, for example, but not limited to goat anti-CHO, affinity purified (Cygnus). A labeled second antibody, or blend of antibodies, specific to the HCPs produced from the cells used to generate the antibody, e.g., anti-CHO HCP Biotinylated, is added, and binds to the HCPs within the sample. In certain embodiments the first and second antibodies are polyclonal antibodies. In certain aspects the first and second antibodies are blends of polyclonal antibodies raised against HCPs. The amount of HCP contained in the sample is determined using the appropriate test based on the label of the second antibody.

HCP ELISA may be used for determining the level of HCPs in an antibody composition, such as an eluate, displacement samples or flow-through fractions obtained using the process described above. The present invention also provides a composition comprising an antibody, wherein the composition has less than 100 ng/mgHCPs as determined by an HCP Enzyme Linked Immunosorbent Assay (“ELISA”).

b. Assaying Charge and Size Variants

In certain embodiments, the levels of product-related substances, such as basic species, acidic species and other variants, in the chromatographic samples produced using the techniques described herein are analyzed. In certain embodiments a CEX-HPLC method is employed. For example, but not by way of limitation, a 4 mm×250 mm analytical Dionex ProPac WCX-10 column (Dionex, Calif.) can be used along with a Shimazhu HPLC system. In certain embodiments, the mobile phases employed in such an assay will include a 10 mM Sodium Phosphate dibasic pH 7.5 buffer (Mobile phase A) and a 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 buffer (Mobile phase B). In certain embodiments, the mobile phases can include a 20 mM MES, pH 6.5 buffer (Mobile phase A) and a 20 mM MES, 500 mM NaCl, pH 6.5 buffer (Mobile phase B). In certain embodiments, the mobile phases can include a 20 mM MES, pH 6.2 buffer (Mobile phase A) and a 20 mM MES, 250 mM NaCl, pH 6.2 buffer (Mobile phase B). In certain embodiments, a binary gradient, for example, but not by way of limitation, a 6% B: 0 min; 6-16% B: 0-20 min; 16-100% B: 20-22 min; 100% B: 22-26 min; 100-6% B: 26-28 min; 6% B: 28-35 min gradient can be used with detection at 280 nm. In certain, non-limiting embodiments, a binary gradient comprising 10% B: 0 min; 10-28% B: 1-46 min; 28-100% B: 46-47 min; 100% B: 47-52 min; 100-10% B: 52-53 min; 10% B: 53-58 min, will be used with detection at 280 nm. In certain embodiments, a binary gradient such as a 1% B: 0-1 min; 1-25% B: 1-46 min; 25-100% B: 46-47 min; 100% B: 47-52 min; 100-1% B: 52-53 min; 1% B: 53-60 min gradient can be used with detection at 280 nm. Quantitation can be based on the relative area percentage of detected peaks. In certain embodiments, the peaks that elute at residence time less than ˜7 min will represent the acidic peaks or AR region. In certain embodiments, all peaks eluting prior to the Main Isoform peak can be summed as the acidic region, and all peaks eluting after the Main peak can be summed as the basic region. In certain embodiments, all peaks eluting prior to the Main Isoform peak (but after, e.g., a 2 min retention time) were summed as the acidic region, and all peaks eluting after the Main peak were summed as the basic region.

In certain embodiments, the levels of aggregates, monomer, and fragments in the chromatographic samples produced using the techniques described herein are analyzed. In certain embodiments, the aggregates, monomer, and fragments are measured using a size exclusion chromatographic (SEC) method for each molecule. For example, but not by way of limitation, a TSK-gel G3000SWxL, 5 μm, 125 Å, 7.8×300 mm column (Tosoh Bioscience) can be used in connection with certain embodiments, while a TSK-gel Super SW3000, 4 μm, 250 Å, 4.6×300 mm column (Tosoh Bioscience) can be used in alternative embodiments. In certain embodiments, the aforementioned columns are used along with an Agilent or a Shimazhu HPLC system. In certain embodiments, sample injections are made under isocratic elution conditions using a mobile phase consisting of, for example, 100 mM sodium sulfate and 100 mM sodium phosphate at pH 6.8, and detected with UV absorbance at 214 nm. In certain embodiments, the mobile phase will consist of 1×PBS at pH 7.4, and elution profile detected with UV absorbance at 280 nm. In certain embodiments, quantification is based on the relative area of detected peaks.

IV. Methods of Treatment

The antibodies, or antigen-binding portions thereof, of the invention comprising a PGPK modification are useful in treating diseases or disorders. In one embodiment, the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof. TNFα has been implicated in the pathophysiology of a wide variety of disorders, including sepsis, infections, autoimmune diseases, transplant rejection and graft-versus-host disease (see e.g., Moeller, A., et al. (1990) Cytokine 2:162-169; U.S. Pat. No. 5,231,024 to Moeller et al.; European Patent Publication No. 260 610 B1 by Moeller, A., et al. Vasilli, P. (1992) Annu. Rev. Immunol. 10:411-452; Tracey, K. J. and Cerami, A. (1994) Annu. Rev. Med. 45:491-503). The present invention provides methods of treating a subject having a disorder in which TNFα activity is detrimental by administering a therapeutically effective amount of an antibody, or antigen-binding portion thereof, of the invention comprising a PGPK modification to the subject, thereby treating the TNFα-associated disease or disorder. In one aspect, the TNFα is human TNFα and the subject is a human subject.

As used herein, the term “a disorder in which TNFα activity is detrimental” is intended to include diseases and other disorders in which the presence of TNFα in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to an excerbation of the disorder. Accordingly, a disorder in which TNFα activity is detrimental is a disorder in which inhibition of TNFα activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of TNFα in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of TNFα in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-TNFα antibody. Disorders in which TNFα activity is detrimental are well known in the art and described in detail in U.S. Pat. No. 8,231,876, the entire contents of which are expressly incorporated herein by reference. Disorders in which TNFα activity is detrimental are also described in “Highlights of Prescribing Information” for HUMIRA® (adalimumab) Injection (Revised January 2008).

In one embodiment, “a disorder in which TNFα activity is detrimental” includes sepsis (including septic shock, endotoxic shock, gram negative sepsis and toxic shock syndrome), autoimmune diseases (including rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis and gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, nephrotic syndrome, multisystem autoimmune diseases, lupus (including systemic lupus, lupus nephritis and lupus cerebritis), Crohn's disease and autoimmune hearing loss), infectious diseases (including malaria, meningitis, acquired immune deficiency syndrome (AIDS), influenza and cachexia secondary to infection), allograft rejection and graft versus host disease, malignancy, pulmonary disorders (including adult respiratory distress syndrome (ARDS), shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibrosis, silicosis, idiopathic interstitial lung disease and chronic obstructive airway disorders (COPD), such as asthma), intestinal disorders (including inflammatory bowel disorders, idiopathic inflammatory bowel disease, Crohn's disease and Crohn's disease-related disorders (including fistulas in the bladder, vagina, and skin; bowel obstructions; abscesses; nutritional deficiencies; complications from corticosteroid use; inflammation of the joints; erythem nodosum; pyoderma gangrenosum; lesions of the eye, Crohn's related arthralgias, fistulizing Crohn's indeterminant colitis and pouchitis), cardiac disorders (including ischemia of the heart, heart insufficiency, restenosis, congestive heart failure, coronary artery disease, angina pectoris, myocardial infarction, cardiovascular tissue damage caused by cardiac arrest, cardiovascular tissue damage caused by cardiac bypass, cardiogenic shock, and hypertension, atherosclerosis, cardiomyopathy, coronary artery spasm, coronary artery disease, valvular disease, arrhythmias, and cardiomyopathies), spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis/spondylitis, enteropathic arthritis, reactive arthritis or Reiter's syndrome, and undifferentiated spondyloarthropathies), metabolic disorders (including obesity and diabetes, including type 1 diabetes mellitus, type 2 diabetes mellitus, diabetic neuropathy, peripheral neuropathy, diabetic retinopathy, diabetic ulcerations, retinopathy ulcerations and diabetic macrovasculopathy), anemia, pain (including acute and chronic pains, such as neuropathic pain and post-operative pain, chronic lower back pain, cluster headaches, herpes neuralgia, phantom limb pain, central pain, dental pain, opioid-resistant pain, visceral pain, surgical pain, bone injury pain, pain during labor and delivery, pain resulting from burns, including sunburn, post partum pain, migraine, angina pain, and genitourinary tract-related pain including cystitis), hepatic disorders (including hepatitis, alcoholic hepatitis, viral hepatitis, alcoholic cirrhosis, al antitypsin deficiency, autoimmune cirrhosis, cryptogenic cirrhosis, fulminant hepatitis, hepatitis B and C, and steatohepatitis, cystic fibrosis, primary biliary cirrhosis, sclerosing cholangitis and biliary obstruction), skin and nail disorders (including psoriasis (including chronic plaque psoriasis, guttate psoriasis, inverse psoriasis, pustular psoriasis and other psoriasis disorders), pemphigus vulgaris, scleroderma, atopic dermatitis (eczema), sarcoidosis, erythema nodosum, hidradenitis suppurative, lichen planus, Sweet's syndrome, scleroderma and vitiligo), vasculitides (including Behcet's disease), and other disorders, such as juvenile rheumatoid arthritis (JRA), endometriosis, prostatitis, choroidal neovascularization, sciatica, Sjogren's syndrome, uveitis, wet macular degeneration, osteoporosis, osteoarthritis, active axial spondyloarthritis (active axSpA) and non-radiographic axial spondyloarthritis (nr-axSpA).

As used herein, the term “subject” is intended to include living organisms, e.g., prokaryotes and eukaryotes. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In specific embodiments of the invention, the subject is a human.

As used herein, the term “treatment” or “treat” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder, as well as those in which the disorder is to be prevented.

The term “dosing” or “dose” or “dosage”, as used herein, refers to the administration of a substance (e.g., an antibody of interest, for example, an anti-TNFα antibody, or antigen-binding portion thereof) to achieve a therapeutic objective (e.g., the treatment or amelioration of a symptom of a disease or disorder).

In one embodiment, the invention provides a method of administering a composition comprising an anti-TNFα antibody, or antigen binding portion thereof, comprising a PGPK modification to a subject such that TNFα activity is inhibited or a disorder in which TNFα activity is detrimental is treated. In one aspect, the TNFα is human TNFα and the subject is a human subject. In one embodiment, the anti-TNFα antibody is adalimumab, also referred to as HUMIRA®.

The compositions comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification can be administered by a variety of methods known in the art. Exemplary routes/modes of administration include subcutaneous injection, intravenous injection or infusion. In certain aspects, a composition comprising an antibody, or antigen-binding portion thereof, containing a PGPK modification may be orally administered. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.

Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. In certain embodiments, it is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit comprising a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.

An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of a composition comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification of the invention is 0.01-20 mg/kg, or 1-10 mg/kg, or 0.3-1 mg/kg. With respect to compositions of the invention comprising an anti-TNFα antibody, or antigen-binding portion thereof, such as adalimumab, comprising a PGPK modification, an exemplary dose is 40 mg every other week. In some embodiments, in particular for treatment of ulcerative colitis or Crohn's disease, an exemplary dose includes an initial dose (Day 1) of 160 mg (e.g., four 40 mg injections in one day or two 40 mg injections per day for two consecutive days), a second dose two weeks later of 80 mg, and a maintenance dose of 40 mg every other week beginning two weeks later. Alternatively, for psoriasis, for example, a dosage can include an 80 mg initial dose followed by 40 mg every other week starting one week after the initial dose.

It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.

V. Pharmaceutical Formulations of Antibodies of the Invention

The present invention further provides preparations and formulations comprising the antibodies (including antibody fragments) comprising a PGPK modification of the present invention. It should be understood that any of the antibodies and antibody fragments described herein, including antibodies and antibody fragments having any one or more of the structural and functional features described in detail throughout the application, may be formulated or prepared as described below. When various formulations are described in this section as including an antibody, it is understood that such an antibody may be an antibody or an antibody fragment having any one or more of the characteristics of the antibodies and antibody fragments described herein. In one embodiment, the antibody is an anti-TNFα antibody, or antigen-binding portion thereof.

In one embodiment, a composition of the invention comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification is formulated with the same or similar excipients and buffers as are present in the commercial adalimumab (HUMIRA®) formulation, as described in the HUMIRA® Prescribing Information, which is expressly incorporated herein by reference. For example, each prefilled syringe of HUMIRA®, which is administered subcutaneously, delivers 0.8 mL (40 mg) of drug product to the subject. Each 0.8 mL of HUMIRA® contains 40 mg adalimumab, 4.93 mg sodium chloride, 0.69 mg monobasic sodium phosphate dehydrate, 1.22 mg dibasic sodium phosphate dehydrate, 0.24 mg sodium citrate, 1.04 mg citric acid monohydrate, 9.6 mg mannitol, 0.8 mg polysorbate 80, and water for Injection, USP. Sodium hydroxide is added as necessary to adjust pH.

In certain embodiments, the antibodies of the invention may be formulated with a pharmaceutically acceptable carrier as pharmaceutical (therapeutic) compositions, and may be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. The term “pharmaceutically acceptable carrier” means one or more non-toxic materials that do not interfere with the effectiveness of the biological activity of the active ingredients. Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents. Such pharmaceutically acceptable preparations may also routinely contain compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration into a human. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being co-mingled with the antibodies of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.

The formulations of the invention are present in a form known in the art and acceptable for therapeutic uses. In one embodiment, a formulation of the invention is a liquid formulation. In another embodiment, a formulation of the invention is a lyophilized formulation. In a further embodiment, a formulation of the invention is a reconstituted liquid formulation. In one embodiment, a formulation of the invention is a stable liquid formulation. In one embodiment, a liquid formulation of the invention is an aqueous formulation. In another embodiment, the liquid formulation is non-aqueous. In a specific embodiment, a liquid formulation of the invention is an aqueous formulation wherein the aqueous carrier is distilled water.

The formulations of the invention comprise an antibody in a concentration resulting in a w/v appropriate for a desired dose. The antibody may be present in the formulation at a concentration of about 1 mg/ml to about 500 mg/ml, e.g., at a concentration of at least 1 mg/ml, at least 5 mg/ml, at least 10 mg/ml, at least 15 mg/ml, at least 20 mg/ml, at least 25 mg/ml, at least 30 mg/ml, at least 35 mg/ml, at least 40 mg/ml, at least 45 mg/ml, at least 50 mg/ml, at least 55 mg/ml, at least 60 mg/ml, at least 65 mg/ml, at least 70 mg/ml, at least 75 mg/ml, at least 80 mg/ml, at least 85 mg/ml, at least 90 mg/ml, at least 95 mg/ml, at least 100 mg/ml, at least 105 mg/ml, at least 110 mg/ml, at least 115 mg/ml, at least 120 mg/ml, at least 125 mg/ml, at least 130 mg/ml, at least 135 mg/ml, at least 140 mg/ml, at least 150 mg/ml, at least 200 mg/ml, at least 250 mg/ml, or at least 300 mg/ml.

In a specific embodiment, a formulation of the invention comprises at least about 100 mg/ml, at least about 125 mg/ml, at least 130 mg/ml, or at least about 150 mg/ml of an antibody of the invention.

In one embodiment, the concentration of antibody, which is included in the formulation of the invention, is between about 1 mg/ml and about 25 mg/ml, between about 1 mg/ml and about 200 mg/ml, between about 25 mg/ml and about 200 mg/ml, between about 50 mg/ml and about 200 mg/ml, between about 75 mg/ml and about 200 mg/ml, between about 100 mg/ml and about 200 mg/ml, between about 125 mg/ml and about 200 mg/ml, between about 150 mg/ml and about 200 mg/ml, between about 25 mg/ml and about 150 mg/ml, between about 50 mg/ml and about 150 mg/ml, between about 75 mg/ml and about 150 mg/ml, between about 100 mg/ml and about 150 mg/ml, between about 125 mg/ml and about 150 mg/ml, between about 25 mg/ml and about 125 mg/ml, between about 50 mg/ml and about 125 mg/ml, between about 75 mg/ml and about 125 mg/ml, between about 100 mg/ml and about 125 mg/ml, between about 25 mg/ml and about 100 mg/ml, between about 50 mg/ml and about 100 mg/ml, between about 75 mg/ml and about 100 mg/ml, between about 25 mg/ml and about 75 mg/ml, between about 50 mg/ml and about 75 mg/ml, or between about 25 mg/ml and about 50 mg/ml.

In a specific embodiment, a formulation of the inventions comprises between about 90 mg/ml and about 110 mg/ml or between about 100 mg/ml and about 210 mg/ml of an antibody.

The formulations of the invention comprising an antibody may further comprise one or more active compounds as necessary for the particular indication being treated, typically those with complementary activities that do not adversely affect each other. Such additional active compound/s is/are suitably present in combination in amounts that are effective for the purpose intended.

The formulations of the invention may be prepared for storage by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers, including, but not limited to buffering agents, saccharides, salts, surfactants, solubilizers, polyols, diluents, binders, stabilizers, salts, lipophilic solvents, amino acids, chelators, preservatives, or the like (Goodman and Gilman's The Pharmacological Basis of Therapeutics, 12th edition, L. Brunton, et al. Remington's Pharmaceutical Sciences, 16th edition, Osol, A. Ed. (1999)), in the form of lyophilized formulations or aqueous solutions at a desired final concentration. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as histidine, phosphate, citrate, glycine, acetate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including trehalose, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN, polysorbate 80, PLURONICS™ or polyethylene glycol (PEG).

The buffering agent may be histidine, citrate, phosphate, glycine, or acetate. The saccharide excipient may be trehalose, sucrose, mannitol, maltose or raffinose. The surfactant may be polysorbate 20, polysorbate 40, polysorbate 80, or Pluronic F68. The salt may be NaCl, KCl, MgCl2, or CaCl2.

The formulations of the invention may include a buffering or pH adjusting agent to provide improved pH control. A formulation of the invention may have a pH of between about 3.0 and about 9.0, between about 4.0 and about 8.0, between about 5.0 and about 8.0, between about 5.0 and about 7.0, between about 5.0 and about 6.5, between about 5.5 and about 8.0, between about 5.5 and about 7.0, or between about 5.5 and about 6.5. In a further embodiment, a formulation of the invention has a pH of about 3.0, about 3.5, about 4.0, about 4.5, about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.5, about 8.0, about 8.5, or about 9.0. In a specific embodiment, a formulation of the invention has a pH of about 6.0. One of skill in the art understands that the pH of a formulation generally should not be equal to the isoelectric point of the particular antibody to be used in the formulation.

Typically, the buffering agent is a salt prepared from an organic or inorganic acid or base. Representative buffering agents include, but are not limited to, organic acid salts such as salts of citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid, or phthalic acid; Tris, tromethamine hydrochloride, or phosphate buffers. In addition, amino acid components can also function in a buffering capacity. Representative amino acid components which may be utilized in the formulations of the invention as buffering agents include, but are not limited to, glycine and histidine. In certain embodiments, the buffering agent is chosen from histidine, citrate, phosphate, glycine, and acetate. In a specific embodiment, the buffering agent is histidine. In another specific embodiment, the buffering agent is citrate. In yet another specific embodiment, the buffering agent is glycine. The purity of the buffering agent should be at least 98%, or at least 99%, or at least 99.5%. As used herein, the term “purity” in the context of histidine and glycine refers to chemical purity of histidine or glycine as understood in the art, e.g., as described in The Merck Index, 13th ed., O'Neil et al. ed. (Merck & Co., 2001).

Buffering agents are typically used at concentrations between about 1 mM and about 200 mM or any range or value therein, depending on the desired ionic strength and the buffering capacity required. The usual concentrations of conventional buffering agents employed in parenteral formulations can be found in: Pharmaceutical Dosage Form: Parenteral Medications, Volume 1, 2nd Edition, Chapter 5, p. 194, De Luca and Boylan, “Formulation of Small Volume Parenterals”, Table 5: Commonly used additives in Parenteral Products. In one embodiment, the buffering agent is at a concentration of about 1 mM, or of about 5 mM, or of about 10 mM, or of about 15 mM, or of about 20 mM, or of about 25 mM, or of about 30 mM, or of about 35 mM, or of about 40 mM, or of about 45 mM, or of about 50 mM, or of about 60 mM, or of about 70 mM, or of about 80 mM, or of about 90 mM, or of about 100 mM. In one embodiment, the buffering agent is at a concentration of 1 mM, or of mM, or of 10 mM, or of 15 mM, or of 20 mM, or of 25 mM, or of 30 mM, or of 35 mM, or of 40 mM, or of 45 mM, or of 50 mM, or of 60 mM, or of 70 mM, or of 80 mM, or of 90 mM, or of 100 mM. In a specific embodiment, the buffering agent is at a concentration of between about 5 mM and about 50 mM. In another specific embodiment, the buffering agent is at a concentration of between 5 mM and 20 mM.

In certain embodiments, the formulation of the invention comprises histidine as a buffering agent. In one embodiment the histidine is present in the formulation of the invention at a concentration of at least about 1 mM, at least about 5 mM, at least about 10 mM, at least about 20 mM, at least about 30 mM, at least about 40 mM, at least about 50 mM, at least about 75 mM, at least about 100 mM, at least about 150 mM, or at least about 200 mM histidine. In another embodiment, a formulation of the invention comprises between about 1 mM and about 200 mM, between about 1 mM and about 150 mM, between about 1 mM and about 100 mM, between about 1 mM and about 75 mM, between about 10 mM and about 200 mM, between about 10 mM and about 150 mM, between about 10 mM and about 100 mM, between about 10 mM and about 75 mM, between about 10 mM and about 50 mM, between about 10 mM and about 40 mM, between about 10 mM and about 30 mM, between about 20 mM and about 75 mM, between about 20 mM and about 50 mM, between about 20 mM and about 40 mM, or between about 20 mM and about 30 mM histidine. In a further embodiment, the formulation comprises about 1 mM, about 5 mM, about 10 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, about 50 mM, about 60 mM, about 70 mM, about 80 mM, about 90 mM, about 100 mM, about 150 mM, or about 200 mM histidine. In a specific embodiment, a formulation may comprise about 10 mM, about 25 mM, or no histidine.

The formulations of the invention may comprise a carbohydrate excipient. Carbohydrate excipients can act, e.g., as viscosity enhancing agents, stabilizers, bulking agents, solubilizing agents, and/or the like. Carbohydrate excipients are generally present at between about 1% to about 99% by weight or volume, e.g., between about 0.1% to about 20%, between about 0.1% to about 15%, between about 0.1% to about 5%, between about 1% to about 20%, between about 5% to about 15%, between about 8% to about 10%, between about 10% and about 15%, between about 15% and about 20%, between 0.1% to 20%, between 5% to 15%, between 8% to 10%, between 10% and 15%, between 15% and 20%, between about 0.1% to about 5%, between about 5% to about 10%, or between about 15% to about 20%. In still other specific embodiments, the carbohydrate excipient is present at 1%, or at 1.5%, or at 2%, or at 2.5%, or at 3%, or at 4%, or at 5%, or at 10%, or at 15%, or at 20%.

Carbohydrate excipients suitable for use in the formulations of the invention include, but are not limited to, monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol) and the like. In one embodiment, the carbohydrate excipients for use in the present invention are chosen from, sucrose, trehalose, lactose, mannitol, and raffinose. In a specific embodiment, the carbohydrate excipient is trehalose. In another specific embodiment, the carbohydrate excipient is mannitol. In yet another specific embodiment, the carbohydrate excipient is sucrose. In still another specific embodiment, the carbohydrate excipient is raffinose. The purity of the carbohydrate excipient should be at least 98%, or at least 99%, or at least 99.5%.

In a specific embodiment, the formulations of the invention may comprise trehalose. In one embodiment, a formulation of the invention comprises at least about 1%, at least about 2%, at least about 4%, at least about 8%, at least about 20%, at least about 30%, or at least about 40% trehalose. In another embodiment, a formulation of the invention comprises between about 1% and about 40%, between about 1% and about 30%, between about 1% and about 20%, between about 2% and about 40%, between about 2% and about 30%, between about 2% and about 20%, between about 4% and about 40%, between about 4% and about 30%, or between about 4% and about 20% trehalose. In a further embodiment, a formulation of the invention comprises about 1%, about 2%, about 4%, about 6%, about 8%, about 15%, about 20%, about 30%, or about 40% trehalose. In a specific embodiment, a formulation of the invention comprises about 4%, about 6% or about 15% trehalose.

In certain embodiments, a formulation of the invention comprises an excipient. In a specific embodiment, a formulation of the invention comprises at least one excipient chosen from: sugar, salt, surfactant, amino acid, polyol, chelating agent, emulsifier and preservative. In one embodiment, a formulation of the invention comprises a salt, e.g., a salt selected from: NaCl, KCl, CaCl2, and MgCl2. In a specific embodiment, the formulation comprises NaCl.

A formulation of the invention may comprise at least about 10 mM, at least about 25 mM, at least about 50 mM, at least about 75 mM, at least about 80 mM, at least about 100 mM, at least about 125 mM, at least about 150 mM, at least about 175 mM, at least about 200 mM, or at least about 300 mM sodium chloride (NaCl). In a further embodiment, the formulation may comprise between about 10 mM and about 300 mM, between about 10 mM and about 200 mM, between about 10 mM and about 175 mM, between about 10 mM and about 150 mM, between about 25 mM and about 300 mM, between about 25 mM and about 200 mM, between about 25 mM and about 175 mM, between about 25 mM and about 150 mM, between about 50 mM and about 300 mM, between about 50 mM and about 200 mM, between about 50 mM and about 175 mM, between about 50 mM and about 150 mM, between about 75 mM and about 300 mM, between about 75 mM and about 200 mM, between about 75 mM and about 175 mM, between about 75 mM and about 150 mM, between about 100 mM and about 300 mM, between about 100 mM and about 200 mM, between about 100 mM and about 175 mM, or between about 100 mM and about 150 mM sodium chloride. In a further embodiment, the formulation may comprise about 10 mM, about 25 mM, about 50 mM, about 75 mM, about 80 mM, about 100 mM, about 125 mM, about 150 mM, about 175 mM, about 200 mM, or about 300 mM sodium chloride.

A formulation of the invention may also comprise an amino acid, e.g., lysine, arginine, glycine, histidine or an amino acid salt. The formulation may comprise at least about 1 mM, at least about 10 mM, at least about 25 mM, at least about 50 mM, at least about 100 mM, at least about 150 mM, at least about 200 mM, at least about 250 mM, at least about 300 mM, at least about 350 mM, or at least about 400 mM of an amino acid. In another embodiment, the formulation may comprise between about 1 mM and about 100 mM, between about 10 mM and about 150 mM, between about 25 mM and about 250 mM, between about 25 mM and about 300 mM, between about 25 mM and about 350 mM, between about 25 mM and about 400 mM, between about 50 mM and about 250 mM, between about 50 mM and about 300 mM, between about 50 mM and about 350 mM, between about 50 mM and about 400 mM, between about 100 mM and about 250 mM, between about 100 mM and about 300 mM, between about 100 mM and about 400 mM, between about 150 mM and about 250 mM, between about 150 mM and about 300 mM, or between about 150 mM and about 400 mM of an amino acid. In a further embodiment, a formulation of the invention comprises about 1 mM, 1.6 mM, 25 mM, about 50 mM, about 100 mM, about 150 mM, about 200 mM, about 250 mM, about 300 mM, about 350 mM, or about 400 mM of an amino acid.

The formulations of the invention may further comprise a surfactant. The term “surfactant” as used herein refers to organic substances having amphipathic structures; namely, they are composed of groups of opposing solubility tendencies, typically an oil-soluble hydrocarbon chain and a water-soluble ionic group. Surfactants can be classified, depending on the charge of the surface-active moiety, into anionic, cationic, and nonionic surfactants. Surfactants are often used as wetting, emulsifying, solubilizing, and dispersing agents for various pharmaceutical compositions and preparations of biological materials. Pharmaceutically acceptable surfactants like polysorbates (e.g., polysorbates 20 or 80); polyoxamers (e.g., poloxamer 188); Triton; sodium octyl glycoside; lauryl-, myristyl-, linoleyl-, or stearyl-sulfobetaine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetyl-betaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-betaine (e.g., lauroamidopropyl); myristamidopropyl-, palmidopropyl-, or isostearamidopropyl-dimethylamine; sodium methyl cocoyl-, or disodium methyl oleyl-taurate; and the MONAQUA™ series (Mona Industries, Inc., Paterson, N.J.), polyethyl glycol, polypropyl glycol, and copolymers of ethylene and propylene glycol (e.g., PLURONICS™, PF68, etc.), can optionally be added to the formulations of the invention to reduce aggregation. In one embodiment, a formulation of the invention comprises Polysorbate 20, Polysorbate 40, Polysorbate 60, or Polysorbate 80. Surfactants are particularly useful if a pump or plastic container is used to administer the formulation. The presence of a pharmaceutically acceptable surfactant mitigates the propensity for the protein to aggregate. The formulations may comprise a polysorbate which is at a concentration ranging from between about 0.001% to about 1%, or about 0.001% to about 0.1%, or about 0.01% to about 0.1%. In other specific embodiments, the formulations of the invention comprise a polysorbate which is at a concentration of 0.001%, or 0.002%, or 0.003%, or 0.004%, or 0.005%, or 0.006%, or 0.007%, or 0.008%, or 0.009%, or 0.01%, or 0.015%, or 0.02%.

The formulations of the invention may optionally further comprise other common excipients and/or additives including, but not limited to, diluents, binders, stabilizers, lipophilic solvents, preservatives, adjuvants, or the like. Pharmaceutically acceptable excipients and/or additives may be used in the formulations of the invention. Commonly used excipients/additives, such as pharmaceutically acceptable chelators (for example, but not limited to, EDTA, DTPA or EGTA) can optionally be added to the formulations of the invention to reduce aggregation. These additives are particularly useful if a pump or plastic container is used to administer the formulation.

Preservatives, such as phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (for example, but not limited to, hexahydrate), alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof can optionally be added to the formulations of the invention at any suitable concentration such as between about 0.001% to about 5%, or any range or value therein. The concentration of preservative used in the formulations of the invention is a concentration sufficient to yield a microbial effect. Such concentrations are dependent on the preservative selected and are readily determined by the skilled artisan.

Other contemplated excipients/additives, which may be utilized in the formulations of the invention include, for example, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antistatic agents, lipids such as phospholipids or fatty acids, steroids such as cholesterol, protein excipients such as serum albumin (human serum albumin (HSA), recombinant human albumin (rHA)), gelatin, casein, salt-forming counterions such as sodium and the like. These and additional known pharmaceutical excipients and/or additives suitable for use in the formulations of the invention are known in the art, e.g., as listed in “Remington: The Science & Practice of Pharmacy”, 21st ed., Lippincott Williams & Wilkins, (2005), and in the “Physician's Desk Reference”, 60th ed., Medical Economics, Montvale, N.J. (2005). Pharmaceutically acceptable carriers can be routinely selected that are suitable for the mode of administration, solubility and/or stability of an antibody, as well known those in the art or as described herein.

It will be understood by one skilled in the art that the formulations of the invention may be isotonic with human blood, wherein the formulations of the invention have essentially the same osmotic pressure as human blood. Such isotonic formulations will generally have an osmotic pressure from about 250 mOSm to about 350 mOSm. Isotonicity can be measured by, for example, using a vapor pressure or ice-freezing type osmometer. Tonicity of a formulation is adjusted by the use of tonicity modifiers. “Tonicity modifiers” are those pharmaceutically acceptable inert substances that can be added to the formulation to provide an isotonity of the formulation. Tonicity modifiers suitable for this invention include, but are not limited to, saccharides, salts and amino acids.

In certain embodiments, the formulations of the present invention have an osmotic pressure from about 100 mOSm to about 1200 mOSm, or from about 200 mOSm to about 1000 mOSm, or from about 200 mOSm to about 800 mOSm, or from about 200 mOSm to about 600 mOSm, or from about 250 mOSm to about 500 mOSm, or from about 250 mOSm to about 400 mOSm, or from about 250 mOSm to about 350 mOSm.

The concentration of any one component or any combination of various components, of the formulations of the invention is adjusted to achieve the desired tonicity of the final formulation. For example, the ratio of the carbohydrate excipient to antibody may be adjusted according to methods known in the art (e.g., U.S. Pat. No. 6,685,940). In certain embodiments, the molar ratio of the carbohydrate excipient to antibody may be from about 100 moles to about 1000 moles of carbohydrate excipient to about 1 mole of antibody, or from about 200 moles to about 6000 moles of carbohydrate excipient to about 1 mole of antibody, or from about 100 moles to about 510 moles of carbohydrate excipient to about 1 mole of antibody, or from about 100 moles to about 600 moles of carbohydrate excipient to about 1 mole of antibody.

The desired isotonicity of the final formulation may also be achieved by adjusting the salt concentration of the formulations. Pharmaceutically acceptable salts and those suitable for this invention as tonicity modifiers include, but are not limited to, sodium chloride, sodium succinate, sodium sulfate, potassium chloride, magnesium chloride, magnesium sulfate, and calcium chloride. In specific embodiments, formulations of the invention comprise NaCl, MgCl2, and/or CaCl2. In one embodiment, concentration of NaCl is between about 75 mM and about 150 mM. In another embodiment, concentration of MgCl2 is between about 1 mM and about 100 mM. Pharmaceutically acceptable amino acids including those suitable for this invention as tonicity modifiers include, but are not limited to, proline, alanine, L-arginine, asparagine, L-aspartic acid, glycine, serine, lysine, and histidine.

In one embodiment the formulations of the invention are pyrogen-free formulations which are substantially free of endotoxins and/or related pyrogenic substances. Endotoxins include toxins that are confined inside a microorganism and are released only when the microorganisms are broken down or die. Pyrogenic substances also include fever-inducing, thermostable substances (glycoproteins) from the outer membrane of bacteria and other microorganisms. Both of these substances can cause fever, hypotension and shock if administered to humans. Due to the potential harmful effects, even low amounts of endotoxins must be removed from intravenously administered pharmaceutical drug solutions. The Food & Drug Administration (“FDA”) has set an upper limit of 5 endotoxin units (EU) per dose per kilogram body weight in a single one hour period for intravenous drug applications (The United States Pharmacopeial Convention, Pharmacopeial Forum 26 (1):223 (2000)). When therapeutic proteins are administered in amounts of several hundred or thousand milligrams per kilogram body weight, as can be the case with antibodies, even trace amounts of harmful and dangerous endotoxin must be removed. In certain specific embodiments, the endotoxin and pyrogen levels in the composition are less then 10 EU/mg, or less then 5 EU/mg, or less then 1 EU/mg, or less then 0.1 EU/mg, or less then 0.01 EU/mg, or less then 0.001 EU/mg.

When used for in vivo administration, the formulations of the invention should be sterile. The formulations of the invention may be sterilized by various sterilization methods, including sterile filtration, radiation, etc. In one embodiment, the antibody formulation is filter-sterilized with a presterilized 0.22-micron filter. Sterile compositions for injection can be formulated according to conventional pharmaceutical practice as described in “Remington: The Science & Practice of Pharmacy”, 2st ed., Lippincott Williams & Wilkins, (2005). Formulations comprising antibodies, such as those disclosed herein, ordinarily will be stored in lyophilized form or in solution. It is contemplated that sterile compositions comprising antibodies are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having an adapter that allows retrieval of the formulation, such as a stopper pierceable by a hypodermic injection needle. In one embodiment, a composition of the invention is provided as a pre-filled syringe.

In one embodiment, a formulation of the invention is a lyophilized formulation. The term “lyophilized” or “freeze-dried” includes a state of a substance that has been subjected to a drying procedure such as lyophilization, where at least 50% of moisture has been removed.

The phrase “bulking agent” includes a compound that is pharmaceutically acceptable and that adds bulk to a lyo cake. Bulking agents known to the art include, for example, carbohydrates, including simple sugars such as dextrose, ribose, fructose and the like, alcohol sugars such as mannitol, inositol and sorbitol, disaccharides including trehalose, sucrose and lactose, naturally occurring polymers such as starch, dextrans, chitosan, hyaluronate, proteins (e.g., gelatin and serum albumin), glycogen, and synthetic monomers and polymers.

A “lyoprotectant” is a molecule which, when combined with a protein of interest (such as an antibody of the invention), significantly prevents or reduces chemical and/or physical instability of the protein upon lyophilization and subsequent storage. Lyoprotectants include, but are not limited to, sugars and their corresponding sugar alcohols; an amino acid such as monosodium glutamate or histidine; a methylamine such as betaine; a lyotropic salt such as magnesium sulfate; a polyol such as trihydric or higher molecular weight sugar alcohols, e.g., glycerin, dextran, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; propylene glycol; polyethylene glycol; PLURONICS™; and combinations thereof. Additional examples of lyoprotectants include, but are not limited to, glycerin and gelatin, and the sugars mellibiose, melezitose, raffinose, mannotriose and stachyose. Examples of reducing sugars include, but are not limited to, glucose, maltose, lactose, maltulose, iso-maltulose and lactulose. Examples of non-reducing sugars include, but are not limited to, non-reducing glycosides of polyhydroxy compounds selected from sugar alcohols and other straight chain polyalcohols. Examples of sugar alcohols include, but are not limited to, monoglycosides, compounds obtained by reduction of disaccharides such as lactose, maltose, lactulose and maltulose. The glycosidic side group can be either glucosidic or galactosidic. Additional examples of sugar alcohols include, but are not limited to, glucitol, maltitol, lactitol and iso-maltulose. In specific embodiments, trehalose or sucrose is used as a lyoprotectant.

The lyoprotectant is added to the pre-lyophilized formulation in a “lyoprotecting amount” which means that, following lyophilization of the protein in the presence of the lyoprotecting amount of the lyoprotectant, the protein essentially retains its physical and chemical stability and integrity upon lyophilization and storage.

In one embodiment, the molar ratio of a lyoprotectant (e.g., trehalose) and antibody molecules of a formulation of the invention is at least about 10, at least about 50, at least about 100, at least about 200, or at least about 300. In another embodiment, the molar ratio of a lyoprotectant (e.g., trehalose) and antibody molecules of a formulation of the invention is about 1, is about 2, is about 5, is about 10, about 50, about 100, about 200, or about 300.

A “reconstituted” formulation is one which has been prepared by dissolving a lyophilized antibody formulation in a diluent such that the antibody is dispersed in the reconstituted formulation. The reconstituted formulation is suitable for administration (e.g., parenteral administration) to a patient to be treated with the antibody and, in certain embodiments of the invention, may be one which is suitable for intravenous administration.

The “diluent” of interest herein is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation, such as a formulation reconstituted after lyophilization. In some embodiments, diluents include, but are not limited to, sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution. In an alternative embodiment, diluents can include aqueous solutions of salts and/or buffers.

In certain embodiments, a formulation of the invention is a lyophilized formulation comprising an antibody of the invention, wherein at least about 90%, at least about 95%, at least about 97%, at least about 98%, or at least about 99% of said antibody may be recovered from a vial upon shaking said vial for 4 hours at a speed of 400 shakes per minute wherein the vial is filled to half of its volume with the formulation. In another embodiment, a formulation of the invention is a lyophilized formulation comprising an antibody of the invention, wherein at least about 90%, at least about 95%, at least about 97%, at least about 98%, or at least about 99% of the antibody may be recovered from a vial upon subjecting the formulation to three freeze/thaw cycles wherein the vial is filled to half of its volume with said formulation. In a further embodiment, a formulation of the invention is a lyophilized formulation comprising an antibody of the invention, wherein at least about 90%, at least about 95%, at least about 97%, at least about 98%, or at least about 99% of the antibody may be recovered by reconstituting a lyophilized cake generated from said formulation.

In one embodiment, a reconstituted liquid formulation may comprise an antibody at the same concentration as the pre-lyophilized liquid formulation.

In another embodiment, a reconstituted liquid formulation may comprise an antibody at a higher concentration than the pre-lyophilized liquid formulation, e.g., about 2 fold, about 3 fold, about 4 fold, about 5 fold, about 6 fold, about 7 fold, about 8 fold, about 9 fold, or about 10 fold higher concentration of an antibody than the pre-lyophilized liquid formulation.

In yet another embodiment, a reconstituted liquid formulation may comprise an antibody of the invention at a lower concentration than the pre-lyophilized liquid formulation, e.g., about 2 fold, about 3 fold, about 4 fold, about 5 fold, about 6 fold, about 7 fold, about 8 fold, about 9 fold or about 10 fold lower concentration of an antibody than the pre-lyophilized liquid formulation.

The pharmaceutical formulations of the invention are preferably stable formulations, e.g., stable at room temperature.

The terms “stability” and “stable” as used herein in the context of a formulation comprising an antibody of the invention refer to the resistance of the antibody in the formulation to aggregation, degradation or fragmentation under given manufacture, preparation, transportation and storage conditions. The “stable” formulations of the invention retain biological activity under given manufacture, preparation, transportation and storage conditions. The stability of the antibody can be assessed by degrees of aggregation, degradation or fragmentation, as measured by HPSEC, static light scattering (SLS), Fourier Transform Infrared Spectroscopy (FTIR), circular dichroism (CD), urea unfolding techniques, intrinsic tryptophan fluorescence, differential scanning calorimetry, and/or ANS binding techniques, compared to a reference formulation. For example, a reference formulation may be a reference standard frozen at −70° C. consisting of 10 mg/ml of an antibody of the invention in PBS.

Therapeutic formulations of the present invention may be formulated for a particular dosage. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the antibody and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an antibody for the treatment of sensitivity in individuals.

Therapeutic compositions of the present invention can be formulated for particular routes of administration, such as oral, nasal, pulmonary, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. By way of example, in certain embodiments, the antibodies (including antibody fragments) are formulated for intravenous administration. In certain other embodiments, the antibodies (including antibody fragments) are formulated for local delivery to the cardiovascular system, for example, via catheter, stent, wire, intramyocardial delivery, intrapericardial delivery, or intraendocardial delivery.

Formulations of the present invention which are suitable for topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required (U.S. Pat. Nos. 7,378,110; 7,258,873; 7,135,180; US Publication No. 2004-0042972; and 2004-0042971).

The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.

Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.

In certain embodiments, antibodies of the invention can be formulated to ensure proper distribution in vivo. For example, the blood-brain bather (BBB) excludes many highly hydrophilic compounds. To ensure that the therapeutic compounds of the invention can cross the BBB (if desired), they can be formulated, for example, in liposomes. For methods of manufacturing liposomes, see, e.g., U.S. Pat. Nos. 4,522,811; 5,374,548; 5,399,331. The liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V. V. Ranade (1989) J. Clin. Pharmacol. 29:685). Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Pat. No. 5,416,016); mannosides (Umezawa et al., (1988) Biochem. Biophys. Res. Commun. 153:1038); antibodies (P. G. Bloeman et al. (1995) FEBS Lett. 357:140; M. Owais et al. (1995) Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al. (1995) Am. J. Physiol. 1233:134), different species of which may comprise the formulations of the invention, as well as components of the invented molecules; p120 (Schreier et al. (1994) J. Biol. Chem. 269:9090); see also K. Keinanen; M. L. Laukkanen (1994) FEBS Lett. 346:123; J. J. Killion; I. J. Fidler (1994) Immunomethods 4:273. In one embodiment of the invention, the therapeutic compounds of the invention are formulated in liposomes; in another embodiment, the liposomes include a targeting moiety. In another embodiment, the therapeutic compounds in the liposomes are delivered by bolus injection to a site proximal to the desired area. When administered in this manner, the composition must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and may be preserved against the contaminating action of microorganisms such as bacteria and fungi. Additionally or alternatively, the antibodies of the invention may be delivered locally to the brain to mitigate the risk that the blood brain barrier slows effective delivery.

In certain embodiments, the therapeutic antibody compositions may be administered with medical devices known in the art. For example, in certain embodiments an antibody or antibody fragment is administered locally via a catheter, stent, wire, or the like. For example, in one embodiment, a therapeutic composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Pat. Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824; 4,596,556. Examples of well-known implants and modules useful in the present invention include: U.S. Pat. No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Pat. No. 4,486,194, which discloses a therapeutic device for administering medicants through the skin; U.S. Pat. No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Pat. No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Pat. No. 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and U.S. Pat. No. 4,475,196, which discloses an osmotic drug delivery system. Many other such implants, delivery systems, and modules are known to those skilled in the art.

The efficient dosages and the dosage regimens for the antibodies of the invention depend on the disease or condition to be treated and can be determined by the persons skilled in the art. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.

a. Alternative Aqueous Formulations

The invention also provides compositions comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification formulated as an aqueous formulation comprising a protein and water, as described in U.S. Pat. No. 8,420,081 and PCT Publication No. WO2012/065072, the contents of each of which are hereby incorporated by reference. In these aqueous formulations, the antibody, or antigen-binding portion thereof, comprising a PGPK modification is stable without the need for additional agents. This aqueous formulation has a number of advantages over conventional formulations in the art, including stability of the protein in water without the requirement for additional excipients, increased concentrations of protein without the need for additional excipients to maintain solubility of the protein, and low osmolality. These also have advantageous storage properties, as the proteins in the formulation remain stable during storage, e.g., stored as a liquid form for more than 3 months at 7° C. or freeze/thaw conditions, even at high protein concentrations and repeated freeze/thaw processing steps. In one embodiment, formulations described herein include high concentrations of antibodies, or antigen-binding portions thereof, comprising a PGPK modification such that the aqueous formulation does not show significant opalescence, aggregation, or precipitation.

In one embodiment, a composition comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification and water is provided, wherein the formulation has certain characteristics, such as, but not limited to, low conductivity, e.g., a conductivity of less than about 2.5 mS/cm, a protein concentration of at least about 10 μg/mL, an osmolality of no more than about 30 mOsmol/kg, and/or the protein has a molecular weight (Mw) greater than about 47 kDa. In one embodiment, the formulation has improved stability, such as, but not limited to, stability in a liquid form for an extended time (e.g., at least about 3 months or at least about 12 months) or stability through at least one freeze/thaw cycle (if not more freeze/thaw cycles). In one embodiment, the formulation is stable for at least about 3 months in a form selected from the group consisting of frozen, lyophilized, or spray-dried.

In one embodiment, the formulation has a low conductivity, including, for example, a conductivity of less than about 2.5 mS/cm, a conductivity of less than about 2 mS/cm, a conductivity of less than about 1.5 mS/cm, a conductivity of less than about 1 mS/cm, or a conductivity of less than about 0.5 mS/cm.

In another embodiment, antibodies, or antigen-binding portions thereof, comprising a PGPK modification of the invention included in the formulation have a given concentration, including, for example, a concentration of at least about 1 mg/mL, at least about 10 mg/mL, at least about 50 mg/mL, at least about 100 mg/mL, at least about 150 mg/mL, at least about 200 mg/mL, or greater than about 200 mg/mL. In another embodiment, the formulation of the invention has an osmolality of no more than about 15 mOsmol/kg.

The aqueous formulations described herein do not rely on standard excipients, e.g., a tonicity modifier, a stabilizing agent, a surfactant, an anti-oxidant, a cryoprotectant, a bulking agent, a lyroprotectant, a basic component, and an acidic component. In other embodiments of the invention, the formulation contains water, one or more proteins, and no ionic excipients (e.g., salts, free amino acids).

In certain embodiments, the aqueous formulations as described herein comprise a composition comprising an antibody, or antigen-binding portion thereof, comprising a PGPK modification having a protein concentration of at least 50 mg/mL and water, wherein the formulation has an osmolality of no more than 30 mOsmol/kg. Lower limits of osmolality of the aqueous formulation are also encompassed by the invention. In one embodiment the osmolality of the aqueous formulation is no more than 15 mOsmol/kg. The aqueous formulation of the invention may have an osmolality of less than 30 mOsmol/kg, and also have a high protein concentration, e.g., the concentration of the protein is at least 100 mg/mL, and may be as much as 200 mg/mL or greater. Ranges intermediate to the above recited concentrations and osmolality units are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.

The concentration of the aqueous formulation as described herein is not limited by the protein size and the formulation may include any size range of proteins. Included within the scope of the invention is an aqueous formulation comprising at least 40 mg/mL and as much as 200 mg/mL or more of a protein, for example, 40 mg/mL, 65 mg/mL, 130 mg/mL, or 195 mg/ml, which may range in size from 5 kDa to 150 kDa or more. In one embodiment, the protein in the formulation of the invention is at least about 15 kD in size, at least about 20 kD in size; at least about 47 kD in size; at least about 60 kD in size; at least about 80 kD in size; at least about 100 kD in size; at least about 120 kD in size; at least about 140 kD in size; at least about 160 kD in size; or greater than about 160 kD in size. Ranges intermediate to the above recited sizes are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.

The aqueous formulation as described herein may be characterized by the hydrodynamic diameter (Dh) of the proteins in solution. The hydrodynamic diameter of the protein in solution may be measured using dynamic light scattering (DLS), which is an established analytical method for determining the Dh of proteins. Typical values for monoclonal antibodies, e.g., IgG, are about 10 nm Low-ionic formulations may be characterized in that the Dh of the proteins are notably lower than protein formulations comprising ionic excipients. It has been discovered that the Dh values of antibodies in aqueous formulations made using the disfiltration/ultrafilteration (DF/UF) process, as described in U.S. Pat. No. 8,420,081 and PCT Publication No. WO2012/065072, using pure water as an exchange medium, are notably lower than the Dh of antibodies in conventional formulations independent of protein concentration. In one embodiment, antibodies in the aqueous formulation as described herein have a Dh of less than 4 nm, or less than 3 nm.

In one embodiment, the Dh of the protein in the aqueous formulation is smaller relative to the Dh of the same protein in a buffered solution, irrespective of protein concentration. Thus, in certain embodiments, protein in an aqueous formulation made in accordance with the methods described herein, will have a Dh which is at least 25% less than the Dh of the protein in a buffered solution at the same given concentration. Examples of buffered solutions include, but are not limited to phosphate buffered saline (PBS). In certain embodiments, proteins in the aqueous formulation of the invention have a Dh that is at least 50% less than the Dh of the protein in PBS in at the given concentration; at least 60% less than the Dh of the protein in PBS at the given concentration; at least 70% less than the Dh of the protein in PBS at the given concentration; or more than 70% less than the Dh of the protein in PBS at the given concentration. Ranges intermediate to the above recited percentages are also intended to be part of this invention, e.g., 55%, 56%, 57%, 64%, 68%, and so forth. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included, e.g., 50% to 80%.

In one aspect, the aqueous formulation includes the antibody, or antigen-binding portion thereof, comprising a PGPK modification at a dosage of about 0.01 mg/kg-10 mg/kg. In another aspect, the dosages of the antibody, or antigen-binding portion thereof, comprising a PGPK modification include approximately 1 mg/kg administered every other week, or approximately 0.3 mg/kg administered weekly. A skilled practitioner can ascertain the proper dosage and regime for administering to a subject.

b. “Solid Unit” Formulations

The present invention also provides stable solid compositions of a protein (preferably a therapeutic protein) and a stabilizer, referred to herein as solid units. These formulations are described, for example, in U.S. Provisional Patent Application 61/893,123, entitled “Stable Solid Protein Compositions and Methods of Making Same”, filed on Oct. 18, 2013, the entire contents of which are expressly incorporated herein by reference. Specifically, it has been discovered that despite having a high proportion of sugar relative to the protein, the solid units of the invention maintain structural rigidity and resist changes in shape and/or volume when stored under ambient conditions, e.g., room temperature and humidity, for extended periods of time. The solid units of the invention remain free-flowing and are able to maintain long-term physical and chemical stability of the protein without significant degradation and/or aggregate formation. The solid units of the invention have many advantages over the art, including that they can be formulated for oral delivery and are easily reconstituted in a diluent, such as water. Because the solid units are readily dissolved, they may be used in dual chamber delivery devices and may be prepared directly in a device for patient use.

As used herein, the term “solid unit,” refers to a composition which is suitable for pharmaceutical administration and comprises a protein, e.g., an antibody or peptide, and a stabilizer, e.g., a sugar. The solid unit has a structural rigidity and resistance to changes in shape and/or volume. In a preferred embodiment, the solid unit is obtained by lyophilizing a pharmaceutical formulation of a therapeutic protein. The solid unit may be any shape, e.g., geometric shape, including, but not limited to, a sphere, a cube, a pyramid, a hemisphere, a cylinder, a teardrop, and so forth, including irregularly shaped units. In one embodiment, the solid unit has a volume ranging from about 1 μl to about 204 μl. In one embodiment, the solid unit is not obtained using spray drying techniques, e.g., the solid unit is not a powder or granule.

As used herein, the phrase “a plurality of solid units” refers to a collection or population of solid units, wherein the collection comprises two or more solid units having a substantially uniform shape, e.g., sphere, and/or volume distribution. In one embodiment, the plurality of solid units is free-flowing.

VI. Kits and Articles of Manufacture Comprising Antibodies of the Invention

Also within the scope of the present invention are kits comprising the antibodies, and antigen-binding portions thereof, of the invention and instructions for use. The term “kit” as used herein refers to a packaged product comprising components with which to administer the antibody, or antigen-binding portion thereof, of the invention for treatment of a disease or disorder. The kit typically comprises a box or container that holds the components of the kit. The box or container is affixed with a label or a Food and Drug Administration approved protocol. The box or container holds components of the invention which are typically contained within plastic, polyethylene, polypropylene, ethylene, or propylene vessels. The vessels can be capped-tubes or bottles. The kit can also include instructions for administering an antibody of the invention.

The kit can further contain one more additional reagents, such as an immunosuppressive reagent, a cytotoxic agent or a radiotoxic agent or one or more additional antibodies of the invention (e.g., an antibody having a complementary activity which binds to an epitope in the TNFα antigen distinct from a first anti-TNFα antibody). Kits typically include a label indicating the intended use of the contents of the kit. The term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit.

The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with a liquid formulation or lyophilized formulation of an antibody or antibody fragment thereof of the invention. In one embodiment, a container filled with a liquid formulation of the invention is a pre-filled syringe. In a specific embodiment, the formulations of the invention are formulated in single dose vials as a sterile liquid. For example, the formulations may be supplied in 3 cc USP Type I borosilicate amber vials (West Pharmaceutical Services—Part No. 6800-0675) with a target volume of 1.2 mL. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.

In one embodiment, a container filled with a liquid formulation of the invention is a pre-filled syringe. Any pre-filled syringe known to one of skill in the art may be used in combination with a liquid formulation of the invention. Pre-filled syringes that may be used are described in, for example, but not limited to, PCT Publications WO05032627, WO08094984, WO9945985, WO03077976, U.S. Pat. Nos. 6,792,743, 5,607,400, 5,893,842, 7,081,107, 7,041,087, 5,989,227, 6,807,797, 6,142,976, 5,899,889, US Patent Publications US20070161961A1, US20050075611A1, US20070092487A1, US20040267194A1, US20060129108A1. Pre-filled syringes may be made of various materials. In one embodiment a pre-filled syringe is a glass syringe. In another embodiment a pre-filled syringe is a plastic syringe. One of skill in the art understands that the nature and/or quality of the materials used for manufacturing the syringe may influence the stability of a protein formulation stored in the syringe. For example, it is understood that silicon based lubricants deposited on the inside surface of the syringe chamber may affect particle formation in the protein formulation. In one embodiment, a pre-filled syringe comprises a silicone based lubricant. In one embodiment, a pre-filled syringe comprises baked on silicone. In another embodiment, a pre-filled syringe is free from silicone based lubricants. One of skill in the art also understands that small amounts of contaminating elements leaching into the formulation from the syringe barrel, syringe tip cap, plunger or stopper may also influence stability of the formulation. For example, it is understood that tungsten introduced during the manufacturing process may adversely affect formulation stability. In one embodiment, a pre-filled syringe may comprise tungsten at a level above 500 ppb. In another embodiment, a pre-filled syringe is a low tungsten syringe. In another embodiment, a pre-filled syringe may comprise tungsten at a level between about 500 ppb and about 10 ppb, between about 400 ppb and about 10 ppb, between about 300 ppb and about 10 ppb, between about 200 ppb and about 10 ppb, between about 100 ppb and about 10 ppb, between about 50 ppb and about 10 ppb, between about 25 ppb and about 10 ppb.

In certain embodiments, kits comprising antibodies of the invention are also provided that are useful for various purposes, e.g., research and diagnostic including for purification or immunoprecipitation of protein of interest from cells, detection of the protein of interest in vitro or in vivo. For isolation and purification of a protein of interest, the kit may contain an antibody coupled to beads (e.g., sepharose beads). Kits may be provided which contain the antibodies for detection and quantitation of a protein of interest in vitro, e.g., in an ELISA or a Western blot. As with the article of manufacture, the kit comprises a container and a label or package insert on or associated with the container. The container holds a composition comprising at least one antibody of the invention. Additional containers may be included that contain, e.g., diluents and buffers, control antibodies. The label or package insert may provide a description of the composition as well as instructions for the intended in vitro or diagnostic use.

The present invention also encompasses a finished packaged and labeled pharmaceutical product. This article of manufacture includes the appropriate unit dosage form in an appropriate vessel or container such as a glass vial, pre-filled syringe or other container that is hermetically sealed. In one embodiment, the unit dosage form is provided as a sterile particulate free solution comprising an antibody that is suitable for parenteral administration. In another embodiment, the unit dosage form is provided as a sterile lyophilized powder comprising an antibody that is suitable for reconstitution.

In one embodiment, the unit dosage form is suitable for intravenous, intramuscular, intranasal, oral, topical or subcutaneous delivery. Thus, the invention encompasses sterile solutions suitable for each delivery route. The invention further encompasses sterile lyophilized powders that are suitable for reconstitution.

As with any pharmaceutical product, the packaging material and container are designed to protect the stability of the product during storage and shipment. Further, the products of the invention include instructions for use or other informational material that advise the physician, technician or patient on how to appropriately prevent or treat the disease or disorder in question, as well as how and how frequently to administer the pharmaceutical. In other words, the article of manufacture includes instruction means indicating or suggesting a dosing regimen including, but not limited to, actual doses, monitoring procedures, and other monitoring information.

Specifically, the invention provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, pre-filled syringe, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of a pharmaceutical agent contained within said packaging material, wherein said pharmaceutical agent comprises a liquid formulation containing an antibody. The packaging material includes instruction means which indicate how that said antibody can be used to prevent, treat and/or manage one or more symptoms associated with a disease or disorder.

The present invention is further illustrated by the following examples which should not be construed as limiting in any way. The contents of all cited references, including literature references, issued patents, and published patent applications, as cited throughout this application are hereby expressly incorporated herein by reference. It should further be understood that the contents of all the figures and tables attached hereto are expressly incorporated herein by reference. The entire contents of the following applications are also expressly incorporated herein by reference: U.S. Provisional Patent Application 61/893,123, entitled “STABLE SOLID PROTEIN COMPOSITIONS AND METHODS OF MAKING SAME”, filed on Oct. 18, 2013; U.S. Provisional Application Ser. No. 61/892,833, entitled “LOW ACIDIC SPECIES COMPOSITIONS AND METHODS FOR PRODUCING THE SAME USING DISPLACEMENT CHROMATOGRAPHY”, filed on Oct. 18, 2013; U.S. Provisional Patent Application 61/892,710, entitled “MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE”, filed on Oct. 18, 2013; U.S. Provisional Patent Application 61/893,068, entitled “LOW ACIDIC SPECIES COMPOSITIONS AND METHODS FOR PRODUCING THE SAME”, filed on Oct. 18, 2013; U.S. Provisional Patent Application 61/893,088, entitled “MODULATED LYSINE VARIANT SPECIES AND METHODS FOR PRODUCING AND USING THE SAME”, filed on Oct. 18, 2013; and U.S. Provisional Patent Application 61/893,131, entitled “PURIFICATION OF PROTEINS USING HYDROPHOBIC INTERACTION CHROMATOGRAPHY”, filed on Oct. 18, 2013.

EXAMPLES Example 1 Analysis of Product-Related Substance Variation of Adalimumab

The production of proteins for biopharmaceutical applications typically involves the use of cell cultures that are known to produce proteins exhibiting varying levels of product-related substance heterogeneity. Such heterogeneity includes, but is not limited to, charge variants such as acidic species and basic species. For example, but not by way of limitation, the charge variants can be separated based on chromatographic residence time. FIG. 1 depicts a non-limiting example of such a division wherein the total acidic species associated with the expression of Adalimumab is divided into a first acidic species region (AR1) and a second acidic species region (AR2), as well as the uncharged product (Lys 0) and the two basic variants, where one C-terminal lysine is present (Lys 1) or both C-terminal lysines are present (Lys 2). The compositions of particular acidic species regions may differ depending on the particular antibody of interest, as well as the particular cell culture, purification, and/or chromatographic conditions employed. As depicted in FIG. 2, the individual charge variants can be resolved from each other.

Example 2 Reduction in Lysine Variation by Treatment with Carboxypeptidase B

Without being bound by theory, it is believed that a C-terminal lysine of antibody will be quickly cleaved enzymatically through the catalysis of carboxypeptidase U. In order to test the susceptibility of antibodies to such enzymatic removal, which would convert Lys 1 and Lys 2 species to Lys 0, a recombinant carboxypeptidase B was incubated with an Adalimumab sample containing variants, including those with terminal lysines. The population of antibody with C-terminal lysine was quickly converted to Lys 0 species as shown in FIG. 3 (compare untreated which includes a population of Lys 1 to CPB treated where essentially the entire population has been converted to Lys 0).

Example 3 Preparation of Modified Anti-TNFα Antibody

In order to minimize enzymatic cleavage of the C-terminal lysine, and thereby reduce product heterogeneity, a modified anti-TNFα antibody was prepared. Specifically, a proline residue was inserted between the C-terminal lysine and the immediately preceding glycine. Thus the C-terminal three amino acids were altered from the original Adalimumab C-terminal sequence, PGK, to a modified C-terminal sequence of PGPK. Without being bound by theory, it is believed that because proline is an imino acid in which the side chain bonds to its backbone nitrogen, the inclusion of a prolines residue at this location will add a kink and rigidity to the peptide backbone and as such will restrict the ability of peptidases to cleave the C-terminal lysine.

Example 4 Physical Properties of Modified Anti-TNFα Antibody

In order to compare the physical properties the modified anti-TNFα antibody prepared above using Adalimumab, the following series of experiments were performed.

Adalimumab and the modified anti-TNFα antibody were incubated in human and rat plasma in order to compare their susceptibility to C-terminal cleavage by plasma proteases. The carboxypeptidase U found in serum coordinates a divalent metal cation in its catalysis. Citrate is an effective anticoagulant but is also a chelator which may strip the cation from the active site and inhibit the enzyme's activity. To address these possible issues, the instant experiment investigates C-terminal processing of either Adalimumab or the modified anti-TNFα antibody in the presence of human plasma with citrate as an anticoagulant, human plasma with heparin as an anticoagulant and mouse plasma with heparin as an anticoagulant. The data is presented in FIG. 6 (Adalimumab) and 7 (the modified anti-TNFα antibody). The modified anti-TNFα antibody was resistant to C-terminal processing when spiked into each of the three different plasma samples. In contrast, Adalimumab, including a sub-population of antibody with C-terminal lysine, was only resistant to processing in the plasma sample that included citrate as an anticoagulant. These data indicate that the chelation of the citrate to the carboxypeptidase U active site inhibits the processing. The two plasma samples with heparin as an anticoagulant have no residual C-terminal lysine constituents. In summary, the data demonstrates that the endogenous carboxypeptidase are capable of processing the normal C-terminus of an antibody but cannot process the modified C-terminus thus allowing it to retain a localized net positive charge.

In a second experiment, mice were immunized at 2 mg/Kg and 5 mg/Kg of the modified anti-TNFα antibody. The mice were then sacrificed and the terminal bleeds were obtained. The samples were passed through an immobilized TNF-alpha column and the eluate was analyzed by LC/MS to evaluate the presence or absence of an intact C-terminal lysine. As depicted in FIG. 7, the modified anti-TNFα antibody retained both C-terminal lysine residues present when analyzed by LC/MS, thus the modified anti-TNFα antibody is able to retain the C-terminal lysines in vivo.

EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

1. An antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9).

2. The antibody, or antigen-binding portion thereof, of claim 1, wherein the antibody, or antigen-binding portion thereof, is an anti-TNFα antibody, or antigen-binding portion thereof.

3. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering a therapeutically effective amount of the antibody, or antigen-binding portion thereof, of claim 2 to the subject, thereby treating the TNFα-associated disease or disorder.

4. The method of claim 3, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

5. The antibody, or antigen-binding portion thereof, of claim 2, wherein the antibody, or antigen-binding portion thereof, comprises the heavy and light chain variable regions of adalimumab.

6. A pharmaceutical composition comprising the antibody, or antigen-binding portion thereof, of claim 5 and a pharmaceutically acceptable carrier.

7. A pharmaceutical composition comprising the antibody, or antigen-binding portion thereof, of claim 2 and a pharmaceutically acceptable carrier.

8. A pharmaceutical composition comprising the antibody, or antigen-binding portion thereof, of claim 1 and a pharmaceutically acceptable carrier.

9. An anti-TNFα antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9).

10. A pharmaceutical composition comprising the antibody, or antigen-binding portion thereof, of claim 9 and a pharmaceutically acceptable carrier.

11. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering a therapeutically effective amount of the antibody, or antigen-binding portion thereof, of claim 9 to the subject, thereby treating the TNFα-associated disease or disorder.

12. The method of claim 11, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

13. An antibody, or antigen-binding portion thereof, comprising the heavy and light chain variable regions of adalimumab and a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9).

14. A pharmaceutical composition comprising the antibody, or antigen-binding portion thereof, of claim 13 and a pharmaceutically acceptable carrier.

15. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering a therapeutically effective amount of the antibody, or antigen-binding portion thereof, of claim 13 to the subject, thereby treating the TNFα-associated disease or disorder.

16. The method of claim 15, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

17. A pharmaceutical composition comprising an antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9) and a pharmaceutically acceptable carrier.

18. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering a therapeutically effective amount of an antibody, or antigen-binding portion thereof, comprising the heavy and light chain variable regions of adalimumab and a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9) to the subject, thereby treating the TNFα-associated disease or disorder.

19. The method of claim 18, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

20. A pharmaceutical composition comprising an anti-TNFα antibody, or antigen-binding portion thereof, comprising a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9) and a pharmaceutically acceptable carrier.

21. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering the pharmaceutical composition of claim 20 to the subject, thereby treating the TNFα-associated disease or disorder.

22. The method of claim 21, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

23. A pharmaceutical composition comprising an antibody, or antigen-binding portion thereof, comprising the heavy and light chain variable regions of adalimumab and a C-terminal heavy chain sequence of proline-glycine-proline-lysine (PGPK) (SEQ ID NO:9), and a pharmaceutically acceptable carrier.

24. A method of treating a subject having a disorder in which TNFα activity is detrimental, the method comprising administering the pharmaceutical composition of claim 23 to the subject, thereby treating the TNFα-associated disease or disorder.

25. The method of claim 24, wherein the disorder in which TNFα activity is detrimental is selected from the group consisting of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's Disease, plaque psoriasis, active axial spondyloarthritis and non-radiographic axial spondyloarthritis.

Referenced Cited
U.S. Patent Documents
4801687 January 31, 1989 Ngo
4877608 October 31, 1989 Lee et al.
4933435 June 12, 1990 Ngo
5045468 September 3, 1991 Darfler
5096816 March 17, 1992 Maiorella
5110913 May 5, 1992 Coan et al.
5118796 June 2, 1992 Prior et al.
5126250 June 30, 1992 McDonough et al.
5231024 July 27, 1993 Moeller et al.
5328985 July 12, 1994 Sano et al.
5429746 July 4, 1995 Shadle et al.
5530101 June 25, 1996 Queen et al.
5545403 August 13, 1996 Page
5545806 August 13, 1996 Lonberg et al.
5545807 August 13, 1996 Surani et al.
5561053 October 1, 1996 Crowley
5569825 October 29, 1996 Lonberg et al.
5625126 April 29, 1997 Lonberg et al.
5633162 May 27, 1997 Keen et al.
5633425 May 27, 1997 Lonberg et al.
5644036 July 1, 1997 Ramage et al.
5654407 August 5, 1997 Boyle et al.
5656272 August 12, 1997 Le et al.
5661016 August 26, 1997 Lonberg et al.
5672347 September 30, 1997 Aggarwal et al.
5672502 September 30, 1997 Birch et al.
5698195 December 16, 1997 Le et al.
5705364 January 6, 1998 Etcheverry et al.
5721121 February 24, 1998 Etcheverry et al.
5770429 June 23, 1998 Lonberg et al.
5789650 August 4, 1998 Lonberg et al.
5795967 August 18, 1998 Aggarwal et al.
5811299 September 22, 1998 Renner et al.
5859205 January 12, 1999 Adair et al.
5876961 March 2, 1999 Crowe et al.
5877293 March 2, 1999 Adair et al.
5929212 July 27, 1999 Jolliffe et al.
5945098 August 31, 1999 Sarno et al.
5976833 November 2, 1999 Furukawa et al.
5994510 November 30, 1999 Adair et al.
6024938 February 15, 2000 Corbo et al.
6036978 March 14, 2000 Gombotz et al.
6048728 April 11, 2000 Inlow et al.
6066719 May 23, 2000 Zapata
6075181 June 13, 2000 Kucherlapati et al.
6090382 July 18, 2000 Salfeld et al.
6113898 September 5, 2000 Anderson et al.
6150584 November 21, 2000 Kucherlapati et al.
6171586 January 9, 2001 Lam et al.
6255458 July 3, 2001 Lonberg et al.
6258562 July 10, 2001 Salfeld et al.
6270766 August 7, 2001 Feldman et al.
6300129 October 9, 2001 Lonberg et al.
6339142 January 15, 2002 Basey et al.
6399381 June 4, 2002 Blum et al.
6406909 June 18, 2002 Shibuya et al.
6410270 June 25, 2002 Strittmatter et al.
6413746 July 2, 2002 Field
6436397 August 20, 2002 Baker et al.
6448380 September 10, 2002 Rathjen et al.
6451983 September 17, 2002 Rathjen et al.
6489447 December 3, 2002 Basey et al.
6498237 December 24, 2002 Rathjen et al.
6509015 January 21, 2003 Salfeld et al.
6528286 March 4, 2003 Ryll
6593458 July 15, 2003 Rathjen et al.
6673986 January 6, 2004 Kucherlapati et al.
6680181 January 20, 2004 Castan
6870034 March 22, 2005 Breece et al.
6872549 March 29, 2005 Van Ness et al.
6890736 May 10, 2005 Reddy et al.
6900056 May 31, 2005 Lee et al.
6914128 July 5, 2005 Salfeld et al.
6924124 August 2, 2005 Singh
6974681 December 13, 2005 McGrew
7070775 July 4, 2006 Le et al.
7084260 August 1, 2006 Lonberg et al.
7122641 October 17, 2006 Vedantham et al.
7189820 March 13, 2007 Ruben
7192584 March 20, 2007 Le et al.
7223394 May 29, 2007 Salfeld et al.
7250165 July 31, 2007 Heavner et al.
7276239 October 2, 2007 Le et al.
7323553 January 29, 2008 Fahrner et al.
7332303 February 19, 2008 Schilling et al.
7390660 June 24, 2008 Behrendt et al.
7429491 September 30, 2008 Luan et al.
7504485 March 17, 2009 Salfeld et al.
7521206 April 21, 2009 Heavner et al.
7521210 April 21, 2009 Knudsen
7541031 June 2, 2009 Salfeld et al.
7588761 September 15, 2009 Salfeld et al.
7645609 January 12, 2010 Follstad
7714112 May 11, 2010 Engstrand et al.
7750129 July 6, 2010 Johansson et al.
7767207 August 3, 2010 Ghayer et al.
7863426 January 4, 2011 Wan et al.
7883704 February 8, 2011 Salfeld et al.
7919264 April 5, 2011 Maksymowych et al.
7947471 May 24, 2011 Knudsen
7972810 July 5, 2011 Crowell et al.
8067182 November 29, 2011 Kelley et al.
8093045 January 10, 2012 Pla et al.
8192951 June 5, 2012 Wang et al.
8197813 June 12, 2012 Salfeld et al.
8206714 June 26, 2012 Salfeld et al.
8209132 June 26, 2012 Bosques et al.
8231876 July 31, 2012 Wan et al.
8361797 January 29, 2013 Osborne et al.
8372400 February 12, 2013 Salfeld et al.
8372401 February 12, 2013 Salfeld et al.
8414894 April 9, 2013 Salfeld et al.
8420081 April 16, 2013 Fraunhofer et al.
8436149 May 7, 2013 Borhani et al.
8663945 March 4, 2014 Pla et al.
8753633 June 17, 2014 Salfeld et al.
20020045207 April 18, 2002 Krummen et al.
20020132299 September 19, 2002 Field
20020187526 December 12, 2002 Ruben et al.
20030012786 January 16, 2003 Teoh et al.
20030049725 March 13, 2003 Heavner et al.
20030096414 May 22, 2003 Ciccarone et al.
20030125247 July 3, 2003 Rosen et al.
20030153735 August 14, 2003 Breece et al.
20030157108 August 21, 2003 Presta
20030161828 August 28, 2003 Abdelghany et al.
20030166869 September 4, 2003 Vedantham et al.
20030175884 September 18, 2003 Umana et al.
20030178368 September 25, 2003 van Reis
20030206898 November 6, 2003 Fischkoff et al.
20030219438 November 27, 2003 Salfeld et al.
20030229212 December 11, 2003 Fahrner et al.
20030235585 December 25, 2003 Fischkoff et al.
20040009172 January 15, 2004 Fischkoff et al.
20040029229 February 12, 2004 Reeves et al.
20040033228 February 19, 2004 Krause et al.
20040038878 February 26, 2004 Tanikawa et al.
20040101939 May 27, 2004 Santora et al.
20040120952 June 24, 2004 Knight et al.
20040126372 July 1, 2004 Banerjee et al.
20040126373 July 1, 2004 Banerjee et al.
20040131614 July 8, 2004 Banerjee et al.
20040132140 July 8, 2004 Satoh et al.
20040136989 July 15, 2004 Banerjee et al.
20040136990 July 15, 2004 Banerjee et al.
20040136991 July 15, 2004 Banerjee et al.
20040151722 August 5, 2004 Banerjee et al.
20040162414 August 19, 2004 Santora et al.
20040166111 August 26, 2004 Kaymakcalan et al.
20040171152 September 2, 2004 Price et al.
20040191243 September 30, 2004 Chen et al.
20040214289 October 28, 2004 deVries et al.
20040219142 November 4, 2004 Banerjee et al.
20050004354 January 6, 2005 Salfeld et al.
20050100965 May 12, 2005 Ghayur et al.
20050123541 June 9, 2005 Heavner et al.
20050175611 August 11, 2005 Mahler et al.
20050249735 November 10, 2005 Le et al.
20050271654 December 8, 2005 Rinderknecht et al.
20050272124 December 8, 2005 Chen et al.
20060009385 January 12, 2006 Hoffman et al.
20060018907 January 26, 2006 Le et al.
20060024293 February 2, 2006 Salfeld et al.
20060083741 April 20, 2006 Hoffman et al.
20060149042 July 6, 2006 Konstantinov et al.
20060153846 July 13, 2006 Krause et al.
20060246073 November 2, 2006 Knight et al.
20060252672 November 9, 2006 Betenbaugh et al.
20060269479 November 30, 2006 Colton et al.
20060287432 December 21, 2006 Christensen et al.
20070003548 January 4, 2007 Heavner et al.
20070004009 January 4, 2007 Dixit et al.
20070041905 February 22, 2007 Hoffman et al.
20070060741 March 15, 2007 Kelley et al.
20070071747 March 29, 2007 Hoffman et al.
20070081996 April 12, 2007 Hoffman et al.
20070110743 May 17, 2007 Drapeau et al.
20070111284 May 17, 2007 Ryll
20070161084 July 12, 2007 Crowell et al.
20070172475 July 26, 2007 Matheus et al.
20070172897 July 26, 2007 Maksymowych et al.
20070184045 August 9, 2007 Doctor et al.
20070184529 August 9, 2007 Etcheverry et al.
20070190057 August 16, 2007 Wu et al.
20070196373 August 23, 2007 Le et al.
20070202051 August 30, 2007 Schuschnig
20070202104 August 30, 2007 Banerjee et al.
20070269463 November 22, 2007 Donovan
20070292442 December 20, 2007 Wan et al.
20070298040 December 27, 2007 Le et al.
20080025976 January 31, 2008 Le et al.
20080112953 May 15, 2008 McAuley et al.
20080118496 May 22, 2008 Medich et al.
20080131374 June 5, 2008 Medich et al.
20080160577 July 3, 2008 Dell'Orco et al.
20080166348 July 10, 2008 Kupper et al.
20080193466 August 14, 2008 Banerjee et al.
20080219952 September 11, 2008 Fischer et al.
20080227136 September 18, 2008 Pla et al.
20080269132 October 30, 2008 Gomes et al.
20080269468 October 30, 2008 Vogel et al.
20080274507 November 6, 2008 Gomes et al.
20080292642 November 27, 2008 Borhani et al.
20080305114 December 11, 2008 Salfeld et al.
20080311043 December 18, 2008 Hoffman et al.
20090017472 January 15, 2009 Stuhlmuller et al.
20090028794 January 29, 2009 Medich et al.
20090053786 February 26, 2009 Kao et al.
20090068172 March 12, 2009 Kaymakcalan et al.
20090068705 March 12, 2009 Drapeau et al.
20090110679 April 30, 2009 Li et al.
20090123378 May 14, 2009 Wong et al.
20090142828 June 4, 2009 Bucciarelli et al.
20090148513 June 11, 2009 Fraunhofer et al.
20090155205 June 18, 2009 Salfeld et al.
20090175857 July 9, 2009 Salfeld et al.
20090202546 August 13, 2009 Harris et al.
20090202557 August 13, 2009 Argiriadi et al.
20090203055 August 13, 2009 Ngantung et al.
20090208500 August 20, 2009 Joly et al.
20090226530 September 10, 2009 Lassner et al.
20090239259 September 24, 2009 Hsieh
20090258018 October 15, 2009 Medich et al.
20090269302 October 29, 2009 Salfeld et al.
20090271164 October 29, 2009 Peng et al.
20090280065 November 12, 2009 Willian et al.
20090291062 November 26, 2009 Fraunhofer et al.
20090304682 December 10, 2009 Hoffman et al.
20090317399 December 24, 2009 Pollack et al.
20100003243 January 7, 2010 Okun et al.
20100016557 January 21, 2010 Salfeld et al.
20100021451 January 28, 2010 Wong
20100034823 February 11, 2010 Borhani et al.
20100040604 February 18, 2010 Salfeld et al.
20100040630 February 18, 2010 Elden et al.
20100135987 June 3, 2010 Hickman et al.
20100136025 June 3, 2010 Hickman et al.
20100160894 June 24, 2010 Julian et al.
20100221823 September 2, 2010 Mccoy et al.
20100256336 October 7, 2010 Yuk et al.
20100278822 November 4, 2010 Fraunhofer et al.
20100297697 November 25, 2010 Ambrosius et al.
20110002935 January 6, 2011 Wan et al.
20110003338 January 6, 2011 Bayer et al.
20110053223 March 3, 2011 Bayer et al.
20110053265 March 3, 2011 Follstad et al.
20110054414 March 3, 2011 Shang et al.
20110081679 April 7, 2011 Jing et al.
20110081700 April 7, 2011 Hasslacher et al.
20110086798 April 14, 2011 Sethuraman et al.
20110097336 April 28, 2011 Wu et al.
20110123544 May 26, 2011 Salfeld et al.
20110130544 June 2, 2011 Ram et al.
20110171227 July 14, 2011 Okun et al.
20110207676 August 25, 2011 Callewaert et al.
20110300151 December 8, 2011 Okun et al.
20120014956 January 19, 2012 Kupper et al.
20120015438 January 19, 2012 Schilling et al.
20120039900 February 16, 2012 Stuhlmuller et al.
20120039908 February 16, 2012 Combs et al.
20120077213 March 29, 2012 Pla et al.
20120107783 May 3, 2012 Julian et al.
20120123688 May 17, 2012 Ramasubramanyan et al.
20120129185 May 24, 2012 Maksymowych et al.
20120171123 July 5, 2012 Medich et al.
20120177596 July 12, 2012 Fischkoff et al.
20120178107 July 12, 2012 Salfeld et al.
20120183997 July 19, 2012 Alley et al.
20120190005 July 26, 2012 Schaub et al.
20120201831 August 9, 2012 Salfeld et al.
20120213792 August 23, 2012 Salfeld et al.
20120219564 August 30, 2012 Salfeld et al.
20120238730 September 20, 2012 Dong et al.
20120244168 September 27, 2012 Salfeld et al.
20120251550 October 4, 2012 Borhani et al.
20120258114 October 11, 2012 Salfeld et al.
20120263731 October 18, 2012 Fraunhofer et al.
20120264920 October 18, 2012 Wang et al.
20120277165 November 1, 2012 Collins et al.
20120282262 November 8, 2012 Okun et al.
20120282270 November 8, 2012 Krause et al.
20120288494 November 15, 2012 Borhani et al.
20120308514 December 6, 2012 Salfeld et al.
20130004507 January 3, 2013 Fischkoff et al.
20130028903 January 31, 2013 Wan et al.
20130065219 March 14, 2013 Tsang et al.
20130084605 April 4, 2013 Zhou et al.
20130096283 April 18, 2013 Khetan et al.
20130115224 May 9, 2013 Salfeld et al.
20130122011 May 16, 2013 Hoffman et al.
20130122018 May 16, 2013 Salfeld et al.
20130156760 June 20, 2013 Fraunhofer et al.
20130195888 August 1, 2013 Wang et al.
20130205604 August 15, 2013 Esenwein et al.
20130243786 September 19, 2013 Banerjee et al.
20130273059 October 17, 2013 Wan et al.
20130280267 October 24, 2013 Wan et al.
20130280274 October 24, 2013 Subramanian et al.
20130309242 November 21, 2013 Wan et al.
20130323261 December 5, 2013 Wan et al.
20130330356 December 12, 2013 Salfeld et al.
20130330357 December 12, 2013 Salfeld et al.
20130336957 December 19, 2013 Wang et al.
20130338344 December 19, 2013 Ramasubramanyan et al.
20130344084 December 26, 2013 Subramanian et al.
20140010820 January 9, 2014 Wang et al.
20140065710 March 6, 2014 Rives et al.
20140072585 March 13, 2014 Herigstad et al.
20140120583 May 1, 2014 Prentice
20140134674 May 15, 2014 Pla et al.
20140134675 May 15, 2014 Pla et al.
20140141007 May 22, 2014 Fraunhofer et al.
20140141008 May 22, 2014 Fraunhofer et al.
20140154270 June 5, 2014 Wang et al.
20140206038 July 24, 2014 Pla et al.
Foreign Patent Documents
1299370 June 2001 CN
1563090 January 2005 CN
3631229 March 1988 DE
0101681 March 1984 EP
0173177 March 1986 EP
0186833 July 1986 EP
0212489 March 1987 EP
0351789 January 1990 EP
0366043 May 1990 EP
0460426 December 1991 EP
0481791 April 1992 EP
0492448 July 1992 EP
0523949 January 1993 EP
0612251 August 1994 EP
0614984 September 1994 EP
0659766 June 1995 EP
0746398 December 1996 EP
0764719 March 1997 EP
0956873 November 1999 EP
0956875 November 1999 EP
1075488 February 2001 EP
1174148 January 2002 EP
1221476 July 2002 EP
1254666 November 2002 EP
1308455 May 2003 EP
1308456 May 2003 EP
1418967 May 2004 EP
1568388 August 2005 EP
1745141 January 2007 EP
1851305 November 2007 EP
2080809 July 2009 EP
2144929 January 2010 EP
2152856 February 2010 EP
2213726 August 2010 EP
2357250 August 2011 EP
2495307 September 2012 EP
2528002 November 2012 EP
2574677 April 2013 EP
2160530 December 1985 GB
2279077 December 1994 GB
7289288 November 1995 JP
WO-90/05144 May 1990 WO
WO-91/02078 February 1991 WO
WO-91/09967 July 1991 WO
WO-92/01047 January 1992 WO
WO-92/11383 July 1992 WO
WO-92/16553 October 1992 WO
WO-93/06213 April 1993 WO
WO-94/02602 February 1994 WO
WO-94/08619 April 1994 WO
WO-94/25585 November 1994 WO
WO-94/26910 November 1994 WO
WO-94/29347 December 1994 WO
WO-9511317 April 1995 WO
WO-95/23813 September 1995 WO
WO-96/33208 October 1996 WO
WO-96/33735 October 1996 WO
WO-96/34096 October 1996 WO
WO-9704801 February 1997 WO
WO-97/13852 April 1997 WO
WO-97/29131 August 1997 WO
WO-98/24884 June 1998 WO
WO-98/24893 June 1998 WO
WO-9823645 June 1998 WO
WO-98/50433 November 1998 WO
WO-9856418 December 1998 WO
WO-99/32605 July 1999 WO
WO-99/57134 November 1999 WO
WO-9957246 November 1999 WO
WO-0003000 January 2000 WO
WO-01-44442 June 2001 WO
WO-0147554 July 2001 WO
WO-01-59069 August 2001 WO
WO-0177362 October 2001 WO
WO-02/12502 February 2002 WO
WO-0212501 February 2002 WO
WO-03045995 June 2003 WO
WO-03/059935 July 2003 WO
WO-03/066662 August 2003 WO
WO-2004008100 January 2004 WO
WO-2004/058944 July 2004 WO
WO-2004058800 July 2004 WO
WO-2005042569 May 2005 WO
WO-2005/082483 September 2005 WO
WO-2006/043895 April 2006 WO
WO-2006045438 May 2006 WO
WO-2006/110277 October 2006 WO
WO-2007/087384 August 2007 WO
WO-2007/117490 October 2007 WO
WO-2008/033517 March 2008 WO
WO-2008-057240 May 2008 WO
WO-2008068879 June 2008 WO
WO-2008087184 July 2008 WO
WO-2008121616 October 2008 WO
WO-2008135498 November 2008 WO
WO-2009023562 February 2009 WO
WO-2009/027041 March 2009 WO
WO-2009058769 May 2009 WO
WO-2009/073569 June 2009 WO
WO-2009135656 November 2009 WO
WO-2010036443 April 2010 WO
WO-2010043703 April 2010 WO
WO-2010122460 October 2010 WO
WO-2010/129469 November 2010 WO
WO-2010127069 November 2010 WO
WO-2011005773 January 2011 WO
WO-2011009623 January 2011 WO
WO-2011-019619 February 2011 WO
WO-2011015926 February 2011 WO
WO-2011024025 March 2011 WO
WO-2011044180 April 2011 WO
WO-2011/073235 June 2011 WO
WO-2011069056 June 2011 WO
WO-2011098526 August 2011 WO
WO-2011110598 September 2011 WO
WO-2011/133886 October 2011 WO
WO-2011127322 October 2011 WO
WO-2011134919 November 2011 WO
WO-2011134920 November 2011 WO
WO-2012019160 February 2012 WO
WO-2012030512 March 2012 WO
WO-2012050175 April 2012 WO
WO-2012051147 April 2012 WO
WO-2012/065072 May 2012 WO
WO-2012062810 May 2012 WO
WO-2012120500 September 2012 WO
WO-2012140138 October 2012 WO
WO-2012145682 October 2012 WO
WO-2012/149197 November 2012 WO
WO-2012147048 November 2012 WO
WO-2012147053 November 2012 WO
WO-2012158551 November 2012 WO
WO-2013/011076 January 2013 WO
WO-2013006461 January 2013 WO
WO-2013006479 January 2013 WO
WO-2013009648 January 2013 WO
WO-2013013013 January 2013 WO
WO-2013-158273 October 2013 WO
WO-2013-158279 October 2013 WO
WO-2013158275 October 2013 WO
WO-2013-176754 November 2013 WO
WO-2013-177115 November 2013 WO
WO-2013-177118 November 2013 WO
WO-2013-181585 December 2013 WO
Other references
  • Cai et al. C-terminal lysine processing of human immunoglobulin G2 heavy chain in vivo. Biotechnol Bioeng. Feb. 2011;108(2):404-12.
  • Abbott Laboratories Press Release, “Abbott Laboratories Receives FDA Approval Earlier Than Expected for Humira (adalimumab) for the Treatment of Rheumatoid Arthritis,” Dec. 31, 2002, pp. 1-4.
  • Averginos, Gab '04 Abstracts—GE Healthcare Life Sciences, “Humira manufacturing: challenges and the path taken”, France, Oct. 3-5, 2004, published 2005, pp. 14-16.
  • Barnes et al., “Stability of Protein Production from Recombinant Mammalian Cells,” Biotechnology and Bioengineering, 81:6, Mar. 20, 2003, pp. 631-639.
  • Boswell et al. “Effects of Charge on Antibody Tissue Distribution and Pharmacokinetics” Bioconjugate Chem.(21) 2153-2163 (2010).
  • Byun, et al. Archives of Biochemistry and Biophysics, “Transport of anti-IL-6 binding fragments into cartilage and the effects of injury,” 532 (2013), pp. 15-22.
  • Canghai, Lu et al.: “A T-flask based screening platform for evaluating and identifying plant hydrolysates for a fed-batch cell culture process”, Cytotechnology, Kluwer Academic Publishers, DO, vol. 55, No. 1, Aug. 18, 2007, pp. 15-29.
  • Chumsae, Chris et al.: “Arginine modifications by methylglyoxal: discovery in a recombinant monoclonal antibody and contribution to acidic species.”, Analytical Chemistry Dec. 3, 2013, vol. 85, No. 23, Dec. 3, 2013, pp. 11401-11409.
  • Clincke, M. et al., “Effect of surfactant pluronic F-68 on CHO cell growth, metabolism, production, and glycosylation of human recombinant IFN-γ in mild operating conditions,” Biotechnol. Prog. 27(1): 181-190, 2011.
  • Du et al., “Chromatographic analysis of the acidic and basic species of recombinant monoclonal antibodies” MAbs, Sep.-Oct. 2012; 4(5):578-85.
  • Feng et al., “Cell Culture Processes for Monoclonal Antibody Production,” mAbs, 2:5, 466-477, Sep./Oct. 2010.
  • Folk et al., “Carboxypeptidase B, Purification and Characterization of the Porcine Enzyme,” J. Biological Chem, 1960, 235:2272-2277.
  • Gramer M Jet al: “Modulation of Antibody Galactosylation Through Feeding of Uridine, Manganese Chloride, and Galactose”,Biotechnology and Bioengineering, Wiley & Sons, Hoboken, NJ, US,vol. 108, No. 7, Jul. 1, 2011, pp. 1591-1682.
  • Gu, X. et al: “Improvement of interferon-gamma sialylation in Chinese hamster ovary cell culture by feeding of N-acetylmannosamine”,Biotechnology and Bioengineering, Wiley & Sons, Hoboken, NJ, US, vol. 58, No. 6, Jun. 20, 1998, pp. 642-648.
  • Hossler P. et al., “Improvement of mammalian cell culture performance through surfactant enabled concentrated feed media,” Biotechnol. Prog. 29(4): 1023-1033, 2013.
  • Humira (adalimumab) prescribing information, Dec. 20, 2002, pp. 1-16.
  • ICH Topic Q6B “Specifications:Test Procedures and Acceptance Criteria for Biotechnological/Biological Products,” Sep. 1999, pp. 1-17.
  • International Search Report from PCT/US2014/024256 dated Jul. 30, 2014, pp. 1-15.
  • Invitation to Pay Additional Fees for International Application No. PCT/US2014/026606, Dated Jul. 8, 2014, pp. 1-8.
  • Luo, Ying et al.: “Development toward rapid and efficient screening for high performance hydrolysate lots in a recombinant monoclonal antibody manufacturing process.”, Biotechnology Progress Jul. 2012, vol. 28, No. 4, Jul. 2012, pp. 1061-1068.
  • Martinelle, K. et al., Cells and Culture, Proceedings of the 20th ESACT Meeting v4 819-822, Jun. 17-20, 2007.
  • Patel, T. P. at al.: “Different culture methods lead to differences in glycosylation of a murine IgG monoclonal antibody”, Biochemical journal, The Biochemical Society, London, GB, vol. 285, No. 3, Jan. 1, 1992, pp. 839-845.
  • Pink, T. et al.: “Regulation of S-layer protein synthesis of Bacillus stearothermophilus PV72 through variation of continuous cultivation conditions”, Journal of Biotechnology, Elsevier Science Publishers, Amsterdam, NL, vol. 50, No. 2, Oct. 1, 1996, pp. 189-200.
  • Rea, J. C. et al.: “Validation of a pH gradient-based ion-exchange chromatography method for high-resolution monoclonal antibody charge variant separations”, Journal of Pharmaceutical and Biomedical Analysis, New York, NY, US, vol. 54, No. 2, Jan. 25, 2011, pp. 317-323.
  • Routier, F. H. et al.: “The glycosylation pattern of a humanized IgGl antibody(D1.3) expressed in CHO cells”, Glycoconjugate Journal, Chapman & Hall, GB, vol. 14, No. 2, Jan. 1, 1997, pp. 201-207.
  • Satoh, Mitsuo et al.: “Non-Fucosylated therapeutic antibodies as next-generation therapeutic antibodies”, Expert opinion on biological therapy, Ashley, London, GB, vol. 6, No. 11, Nov. 1, 2006, pp. 1161-1173.
  • Schiestl et al. “Acceptable changes in quality attributes of glycosylated biopharmaceuticals” Nature Biotechnology, 29(4), 310-312 (2011).
  • Senczuk et al. “Hydrophobic interaction chromatography in dual salt system increases protein binding capacity” Biotechnology and Bioengineering, 103(5), 930-935 (2009).
  • Sundaram et al., “An innovative approach for the characterization of the isoforms of a monoclonal antibody product,” Mabs, 3(6):505-512, 2011.
  • Sung, Hyun Kim et al.: “Development of serum-free medium supplemented with hydrolysates for the production of therapeutic antibodies in CHO cell cultures using design of experiments”, Applied Microbiology and Biotechnology, Springer, Berlin, DE, vol. 83, No. 4, Mar. 6, 2009, pp. 639-648.
  • Wolff et al., “The Kinetics of Carboxypeptidase B Activity,” J. Biological Chem, 1962, 237:3094-3099.
  • Wong N.S.C. et al: “An investigation of intracellular glycosylation activities in CHO cells: Effects of nucleotide sugar precursor feeding” Biotechnology and Bioengineering, vol. 187, No. 2, Oct. 1, 2010, pp. 321-336.
  • Worthington Biochemical Corporation, porcine pancreas carboxypeptidase B, one page, Feb. 25, 2012.
  • “Genentech unveils production capacity hikes,” in-Pharma Technologist.com Jun. 28, 2005, pp. 1-2.
  • “Memorandum in Support of Centocor's Motion for Summary Judgment No. 1 that All Asserted Claims Are Invalid for Lack of Written Description”, dated Aug. 1, 2013 and submitted by defendant in Civil Action No. 09-40089-FDS.
  • “Memorandum in Support of Centocor's Motion for Summary Judgment No. 2 that All Asserted Claims Are Invalid for Lack of Enablement”, dated Aug. 1, 2013 and submitted by defendant in Civil Action No. 09-40089-FDS.
  • “Memorandum in Support of Centocor's Motion for Summary Judgment No. 4 that Claims Encompassing Non-recombinant Human Antibodies Are Invalid for Failing to Meet the Requirements of 35 U.S.C. §112”, dated Aug. 1, 2013 and submitted by defendant in Civil Action No. 09-40089-FDS.
  • “Memorandum in Support of Centocor's Motion No. 3 for Summary Judgment that the 394 and 031 Patents Are Invalid for Under 35 U.S.C. §102(f) for Failing to Name the Proper Inventors”, dated Aug. 1, 2013 and submitted by defendant in Civil Action No. 09-40089-FDS.
  • “Memorandum in Support of Centocor's Motion No. 6 for Summary Judgment that References Dated Before Feb. 10, 1997 Qualify as Prior Art to the 394 and 031 Patents”, dated Aug. 1, 2013 and submitted by defendant in Civil Action No. 09-40089-FDS.
  • “Plaintiffs' Memorandum in Support of Their Motion for Partial Summary Judgment”, dated Aug. 1, 2013 and submitted by plaintiff in Civil Action No. 09-40089-FDS.
  • “Plaintiffs' Rule 56.1 Statement of Undisputed Material Facts in Support of Their Motion for Partial Summary Judgment”, dated Aug. 1, 2013 and submitted by plaintiff in Civil Action No. 09-40089-FDS.
  • Abraham, E., et al., “Efficacy and Safety of Monoclonal Antibody to Human Tumor Necrosis Factor α in Patients with Sepsis Syndrome,” JAMA, vol. 273(12):934-941 (1995).
  • Adams. et al. J. Am. Acad. Dermatol 2004;51 :660-2.
  • Altamirano, C., et al., “Strategies for fed batch cultivation of t-PA producing CHO cells: substitution of glucose and glutamine and rational design of culture medium”, J. Biotechn. 110:171-179, 2004.
  • Antes et al. “Analysis of lysine clipping of a humanized Lewis-Y specific IgG antibody and its relation to Fc-mediated effector function” Journal of Chromatography B:Biomedical Sciences and Applications, Elsevier, Amsterdam, NL, vol. 852, No. 1-2, May 31, 2007, 250-256.
  • Avgerinos et al. (GAb '04 Abstracts—GE Healthcare Life Sciences, France Oct. 3-5, 2004, pp. 15-16 published 2005).
  • Azevedo et al., “Integrated Process for the Purification of Antibodies Combining Aqueous Two-Phase Extraction, Hydrophobic Interaction Chromatography and Size-Exclusion Chromatography”, Journal of Chromatography (2008) 1213(2): 154-161.
  • Ballez, J.S. et al., “Plant protein hydrolysates support CHO-320 cells proliferation and recombinant IFN-[gamma] production in suspension and inside microcarriers in protein-free media”, Cytotechnology 44:3, 103-114, 2004.
  • Barbuto, J. et al. “Production of Neutralizing Antibodies to Tumor Necrosis Factor by Human Tumor-Infiltrating B Lymphocytes” Proc. Am. Assoc. Cancer Res,. 34:487, Abstr. 2904 (1993).
  • Bendtzen, K. et al. “Auto-antibodies to IL-1α and TNFα in Normal Individuals and in Infectious and Immunoinflammatory Disorders” The Physiological and Pathological Effects of Cytokines, 447-52 (1990).
  • Biblia, T.A. et al., “In Pursuit of the Optimal Fed-Batch Process for Monoclonal Antibody Production”, Biotechnol. Prog 11(1):1-13, Jan.-Feb. 1995.
  • Birch, Jr. et al., “Antibody production”, Adv. Drug Delivery Reviews 58:671-685, 2006.
  • Blaker, GJ, et al., “The Glucose, Insulin and Glutamine Requirements of Suspension Cultures of HeLa Cells in a Defined Culture Medium”, J. Cell Sci. 9:529-537, 1971.
  • Boekstegers, P., et al., “Repeated administration of a F(ab′)2 fragment of an anti-tumor necrosis factor alpha monoclonal antibody in patients with severe sepsis: effects on the cardiovascular system and cytokine levels,” Shock, vol. 1(4):237-245 (1994).
  • Bollati-Fogolin M., et al., “Temperature Reduction in Cultures of hGM-CSF-expressing CHO Cells: Effects on Productivity and Product Quantity”, Biotechnol. Prog. 21:17-21, 2005.
  • Bonafede et al. “Cost per treated patient for etanercept, adalimumab, and infliximab across adult indications: a claims analysis” Advances in Therapy, Springer Healthcare Communications, Heidelberg, vol. 29, No. 3, Mar. 9, 2012, 234-249.
  • Boyle, P. et al. “A Novel Monoclonal Human IgM Autoantibody which Binds Recombinant Human and Mouse Tumor Necrosis Factor-α” Cell. Immunol., 152:556-68 (1993).
  • Boyle, P. et al. “The B5 Monoclonal Human Autoantibody Binds to Cell Surface TNFα on Human Lymphoid Cells and Cell Lines and Appears to Recognize a Novel Epitope” Cell. Immunol., 152:569-81 (1993).
  • Brekke, O. et al., “Therapeutic Antibodies for Human Diseases at the Dawn of the Twenty-first Century,” Nature, vol. 2:52-62 (2002).
  • Brorson et al., “Bracketed Generic Inactivation of Rodent Retroviruses by Low pH Treatment; for Monoclonal Antibodies and Recombinant Proteins,” Biotechnology and Bioengineering,; vol. 82(3): 321-329 (2003).
  • Bruggemann et al., “Production of human antibody repertoires in transgenic mice” Cur. Op. Biotechnol. *;455-458 (1997).
  • Bruggemann, M., Neuberger, M.S., “Strategies for expressing human antibody repertoires in transgenic mice,” Immunol. Today 17:391-397 (1996).
  • Cai B, et al. “C-Terminal Lysine Processing of Human; Immunoglobulin G2 Heavy Chain in Vivo” Biotechnol. Bioeng. 2011;108: 404-412.
  • Cambridge Antibody Technology, advertisement of phage display services, Science vol. 253, No. 5018 (1991).
  • Carter et al., “Humanization of an anti-p185HER2 antibody for human cancer therapy,” Proc. Nat. Acad. Sci89:4285-4289 (1992).
  • Chang KH, et al., “N-Acetylcysteine Increases the Biosynthesis of Recombinant EPO in Apoptotic Chinese Hamster Ovary Cells”, Free Radic Res. 30(2):85-91, 1999.
  • Charter, Edward A., “A New Process for the Separation and Purification of Egg Yolk; Antibodies,” BASc., The University of British Columbia; A Thesis; Apr. 1993.
  • Choo et al. “High-level production of a monoclonal antibody in murine myeloma cells by perfusion culture using a gravity settler” Biotechnology Progress, vol. 23, No. 1, Jan. 1, 2007, 225-231.
  • Chow, A. et al. “Effect of monoclonal antibody on human tumor necrosis factor (TNF MAb) on TNFα, IL-1β, and IL-6 levels in patients with sepsis syndrome” Clinical Research, 42:2 299A (1994).
  • Chua, FKF et al., “Hyper-stimulation of monoclonal antibody production by high osmolarity stress in eRDF medium”, J. Biotechnology 37(3):265-275, Nov. 15, 1994.
  • Chung et al., “Utilization of Lysozyme Charge Ladders to Examine the Effects of Protein Surface; Charge Distribution on Bindinq Affinity in Ion Exchange Systems,” Lanqmuir 26(2): 759-768 (2010).
  • Chung et al., “Cetuximab-Induced Anaphylaxis and IgE Specific for Galactose-α-1, 3-Galactose”, N. Engl. J. Med., 358:11, pp. 1109-1117 (2008).
  • Cleland, J. et al., “A Specific Molar Ratio of Stabilizer to Protein is Required for Storage Stability of a Lyophilized Monoclonal Antibody,” Journal of Pharmaceutical Sciences, vol. 90(3):310-321 (2001).
  • Cohen, J., et al., “Intersept: An international, multicenter, placebo-controlled trial of monoclonal anitbody to human tumor necrosis factor-α in patients with sepsis,” Crit Care Med, vol. 24(9):1431-1440 (1996).
  • Cox, J. et al. “A directory of human germ-line Vκ segments reveals a strong bias in their usage” Eur. J. Immunol., 24(2):827-36 (1994).
  • Cromwell (GAB'04 Abstracts—GE Healthcare Life Sciences, Franc Oct. 3-5, 2004, pp. 17-18 published 2005).
  • Daugherty, et al. Formulation and Delivery Issues for Monoclonal Antibody Therapeutics. Advanced Drug Delivery Reviews, 2006. vol. 58, pp. 686-706.
  • Davies et al., “Antibody VH domains as small recognition units.” Biotechnology, 13:475-479 (1995).
  • Department of Surgery, University of Toronto, Annual Report (1998-1999)(348 pages).
  • DePhillips et al., “Determinants of protein retention characteristics on cation-exchange adsorbents,”; Journal of Chromatograph A, 933:57-72 (2001).
  • deZongotita et al., “Phosphate feeding improves high-cell-concentration NS0 myeloma cell culture performance for monoclonal antibody production” Biotechnology and Bioengineering. 2000, 69: 566-576.
  • Dick et al: “C-terminal lysine variants in fully human monoclonal antibodies: Investigation of test methods; and possible causes”, Biotechnology and Bioengineering, vol. 100, No. 6, Aug. 15, 2008, pp. 1132-1143.
  • Dolezal, et al., “Escherichia coli Expression of a Bifunctional Fab-peptide Epitope Reagent for the Rapid Diagnosis of HIV-1 and HIV-2”, Immunotechnology, 1:197-209 (1995).
  • Doring, E., “Identification and Characterization of a TNFa Antagonist Derived from a Monoclonal Antibody” (1994) Mol. Immunol .31(14): 1059-1067.
  • Elliot et al., “Repeated therapy with monoclonal antibody to tumour necrosis factor α (cA2) in patients with rheumatoid arthritis” (1994) Lancet, 344:1125-1127.
  • Elliot, “Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to tumor necrosis factor α” (1993) Arthritis & Rheumatism, 36(12):1681-1690.
  • Emery, P. “Adalimumab therapy: Clinical findings and implications for integration into clinical guidelines for rheumatoid arthritis.” Drugs of Today, 41(3): p. 155-153. (2005).
  • Erbitux (cetuximab) label, Revised Aug. 2013.
  • Ewert et al., “Biophysical Properties of Human Antibody Variable Domains,” J. Mol. Biol. 324: 531-553 (2003).
  • Exhibit dated Aug. 1, 2013 and cited by defendant in Civil Action No. 09-40089-FDS providing the Jun. 23, 2009 trial transcript of the PM session in the matter of Centocor, et al. v. Abbott Laboratories.
  • Exhibit dated Aug. 1, 2013 and cited by defendant in Civil Action No. 09-40089-FDS providing the trial transcript in the matter of Abbott Laboratories, et al. v. The Mathilda and Terrance Kennedy Institute, S.D.N.Y.
  • Exhibit dated Aug. 1, 2013 and cited by plaintiff in Civil Action No. 09-40089-FDS providing excerpts from the File History of U.S. Appl. No. 12/578,487.
  • Exhibit dated Aug. 1, 2013 and cited by defendant in Civil Action No. 09-40089-FDS providing the Jun. 24, 2009 trial transcript of the AM session in the matter of Centocor, et al. v. Abbott Laboratories, E.D. TX.
  • Exhibit dated Aug. 1, 2013 and cited by defendant in Civil Action No. 09-40089-FDS providing the Sep. 20, 2012 Day 8 trial transcript in the matter of Abbott v. Centocor Ortho Biotech Inc., D. MA.
  • Exhibit dated Aug. 1, 2013 and cited by plaintiff in Civil Action No. 09-40089-FDS providing Declaration by Jochen Salfeld, dated Jan. 17, 2013.
  • FDA Package insert for Adalimumab, Sep. 26, 2003, pp. 1-18.
  • Feldmann, “Anti-TNF-alpha Therapy of Rheumatoid Arthritis: What Have We Learned?” (2001) Annu. Rev. Immunol., 19:163-196.
  • Figini, “In Vitro assembly of Repertoires of Antibody Chains on the Surface of Phage by Renaturation” (1994) J. Mol. Biol., 239:68-78.
  • Fishwild et al., “High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice” (1996) Nature Biotechnology, 14:845-851.
  • Fomsgaard, “Auto-antibodies to Tumor Necrosis Factor α in Healthy Humans and Patients with Inflammatory Diseases and Gram-Negative Bacterial Infections” (1989) Scand. J. Immunol. 30:219-23.
  • Foote, J., “Antibody framework residues affecting the conformation of the hypervariable loops” (1992) J. Mol .Biol., 224(2):487-499.
  • Freitag et al., “Displacement chromatography in biotechnological downstream processing,” J. Chromatography, (1995) 691(1):101-112.
  • Gagnon et al., “A Systematic Approach to the Purification of Monoclonal Antibodies,” LC-GC 11 (1):26-34 (1993).
  • Gatto, B. “Biologics targeted at TNF: design, production and challenges”, Reumatismo 58(2):94-103, 2006.
  • Genbank Entry for CHO Cathepsin L., EGW13555, Aug. 25, 2011, pp. 1-2.
  • Ghaderi, et al., “Implications of the Presence of N-glycolylneuraminic acid in Recombinant Therapeutic Glycoproteins”, Nature Biotechnology, 28(8):863-868 (2010).
  • Ghaderi, et al., “Production platforms for biotherapeutic glycoproteins. Occurrence, impact, and challenges of non-human sialylation”, Biotechnology and Genetic Engineering Reviews, 28:147-176 (2012).
  • Gonzalez et al. “Purification of Lactic Acid from Fermentation Broths by Ion-Exchange Resins” Ind. Eng. Chem. Res. 45:3243 (2006).
  • Graf et al., “Ion exchange resins for the purification of monoclonal antibodies from animal cell culture” Bioseparation 4 (1) :7-20 (Feb. 1994). ;4 (1) :7-20 (Feb. 1994).
  • Gram et al., “In vitro selection and affinity maturation of antibodies from a naive combinatorial immunoglobulin library” (1992) PNAS, 89:3576-3580.
  • Gramer et al., “Glycosidase Activities of the 293 and NS0 Cell Lines, and of an Antibody-Producing Hybridoma Cell Line”, Biotechnology and Bioengineering, 43:423-428 (1994).
  • Green et al., “Antigen-specific human monoclonal antibodies from mice engineered with human Ig heavy and light chain YACs” (1994) Nature Genetics, 7:13-21.
  • Griffiths et al., “Isolation of high affinity human antibodies directly from large synthetic repertoires” (1994) EMBO J., 13:3245-3260.
  • Griffiths, “Human anti-self antibodies with high specificity from phage display libraries” (1993) The EMBO J. 12(2):725-34.
  • Grunberg, J. et al., “High-Yield Production of Recombinant Antibody Fragments in HEK-293 Cells Using Sodium Butyrate”, BioTechniques 34(5):968-972, May 2003.
  • Han, Kyu Oh et al., “Effect of N-Acetylcystein on Butyrate-Treated Chinese Hamster Ovary Cells to Improve the Production of Recombinant Human Interferon-β-1a”, Biotechnol. Prog. 21(4):1154-1164, 2005.
  • Harding et al., “Class switching in human immunoglobulin transgenic mice” (1995) Ann. NY Acad. Sci., 764:536-547.
  • Harlow and Lane, Antibodies A Laboratory Manual, Purification of Antibodies by using a; Deae-matrix (Batch), Storing and Purifying Antibodies; Chapter 8: 302-303 (1988).
  • Harris et al. “Processing of C-terminal lysine and argnine residues of proteins isolated from mammalian cell culture” Journal of Chromatography, (1995) 705; 129-123.
  • Harrison et al., “Protein N-Glycosylation in the Baculovirus-Insect Cell Expression System and; Engineering of Insect Cells to Produce “Mammalianized” Recombinant Glycoproteins,” Advances in; Virus Research, 68:159-191 (2006).
  • Hawkins, “Selection of Phage Antibodies by Binding Affinity Mimicking Affinity Maturation” (1992) J. Mol. Biol., 226:889-896.
  • Heidemann, R. et al., “The use of peptones as medium additives for the production of a recombinant therapeutic protein in high density perfusion cultures of mammalian cells”, Cytotechnology 32:157-167, 2000.
  • Helms et al., “Destabilizing loop swaps in the CDRs of an immunoglobulin VL domain,” Protein; Science 4:2073-2081 (1995).
  • Hiatt et al., “Characterization and Applications of Antibodies Produced in Plants”, Intern. Rev. Immunol., 10:139-152 (1993).
  • Hiatt et al., “Production of Antibodies in Transgenic Plants”, Nature, 342:76-78 (1989).
  • Hillgren, A. et al., “Protection mechanism of Tween 80 during freeze-thawing of a model protein LDH,” International Journal of Pharmaceutics, vol. 237:57-69 (2002).
  • Hokke et al., “Sialylated Carbohydrate Chains of Recombinant Human Glycoproteins Expressed in Chinese Hamster Ovary Cells Contain Traces of N-glycolylneuraminic acid”, FEBS, 275:9-14 (1990).
  • Holler, “Modulation of Acute Graft-Versus-Host Disease After Allogeneic Bone Marrow Transplantation by Tumor Necrosis Factor-alpha (TNF-alpha) Release in the Course of Pretransplant Conditioning: Role of Conditioning Regimens and Prophylactic Application of a Monoclonal Antibody Neutralizing Human TNF-alpha (MAK 195F)” (1995) Blood, 86(3):890-899.
  • Holt, L. et al., “Domain antibodies: proteins for therapy,” Trends in Biotechnology, vol. 21(11):484-490 (2003).
  • Hoogenboom et al., “By-passing immunisation : Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro” (1992) J. Mol. Biol., 227:381-388.
  • Hoogenboom, “Converting rodent into human antibodies by guided selection” (1996) Antibody Engineering, Oxford University Press, pp. 169-185.
  • Horvath et al: “Characterization of a Monoclonal Antibody Cell Culture Production Process Using a Quality by; Design Approach”, Molecular Biotechnology, vol. 45, No. 3, Jul. 1, 2010, pp. 203-206.
  • http://www.cygnustechnologies.com/productdetail/host-cell-protein-antibodies/anti-cho-h . . . Cygnus Technologies, Anti-CHO HCP (Apr. 18, 2012).
  • Huang et al. “Effects of anti-TNF monoclonal antibody infusion in patients with hairy cell leukaemia” (1992) Br. J. Haematol., 81(2):231-234.
  • Hui et al., “Recovery and purification process development for monoclonal antibody production,” MABS (2010) 2(5):480-499.
  • Humira (adalimumab) label, Revised Sep. 2013.
  • Huse, “Generation of a large combinatorial library of the immunoglobulin repertoire in phage lambda” (1989) Science, 246:1275-81.
  • International Preliminary Report on Patentability for Application No. PCT/US07/08359, dated Dec. 12, 2011.
  • International Preliminary Report on Patentability for Application No. PCT/US2011/060388, dated May 30, 2012.
  • International Search Report and Written Opinion for Application No. PCT/US2008/085066, dated May 12, 2009.
  • International Search Report and Written Opinion for Application No. PCT/US2010/033387, dated Aug. 7, 2012.
  • International Search Report and Written Opinion for Application No. PCT/US2013/031380, dated Feb. 5, 2014.
  • International Search Report and Written Opinion for Application No. PCT/US2013/041954, dated Dec. 17, 2013.
  • International Search Report and Written Opinion for Application No. PCT/US2013/041958, dated Dec. 17, 2013.
  • International Search Report and Written Opinion for Application No. PCT/US2013/065720, dated Dec. 16, 2013.
  • International Search Report and Written Opinion for Application No. PCT/US2013/065797, dated Nov. 26, 2013.
  • International Search Report for Application No. PCT/IB03/04502, dated May 26, 2004.
  • International Search Report for Application No. PCT/US2011/060388 dated May 30, 2012.
  • International Search Report for Application No. PCT/US2013/031352, Dated Apr. 25, 2013.
  • International Search Report for Application No. PCT/US2013/031389, Dated Jun. 3, 2013.
  • International Search Report for Application No. PCT/US2013/031485, Dated Jun. 25, 2013.
  • International Search Report for Application No. PCT/US2013/031681, Dated Jun. 14, 2013.
  • Invitation to Pay Additional Fees for International Application No. PCT/US2013/031380, Dated Nov. 28, 2013.
  • Jakobovits, A., “Production of fully human antibodies by transgenic mice” (1995) Curr. Op. Biotechnol., 6:561-566.
  • Jespers, “Guiding the Selection of Human Antibodies from Phage Display Repertoires to a Single Epitope of an Antigen” (1994) Bio/Technology, 12:899-903.
  • Johnson et al. (Archives of Biochemistry and Biophysics 444 (2005) 7-14).
  • Kalyanpur, M., “Downstream Processing in the Biotechnology Industry” Molecular Biotechnology, vol. 22:87-98 (2002).
  • Karampetsou et al. (Q J Med 2010; 103:917-928).
  • Kaschak et al: “Characterization of the basic charge variants of a human IgGl: Effect of copper concentration in cell culture media”, MABS, vol. 3, No. 6, Nov. 1, 2011, pp. 577-583.
  • Kazuaki, F. et al., “Enhancement of productivity of recombinant α-amidating enzyme by low temperature culture”, Cytotechnology 31:85-94, 1999.
  • Kempeni, “Update on D2E7: a fully human anti-tumour necrosis factor -alpha monoclonal antibody” (2000) Ann. Rheum. Dis., 59(Suppl. I)144-145.
  • Kempeni, J, “Preliminary results of early clinical trials with the fully human anti-TNFα monoclonal antibody D2E7”, Ann. Rheum. Dis., 1999, pp. 170-172, vol. 58, (Suppl. I).
  • Kempf, C, et al. “Virus inactivation during production of intravenous immunoglobulin.” Transfusion 1991; vol. 31: p. 423-427.
  • Khawli et al, “Charge variants in IgGl: Isolation, characterization, in vitro binding properties and pharmacokinetics in rats”, MABS, vol. 2, No. 6, Nov. 1, 2010, pp. 613-624.
  • Kim, NS. et al., “Inhibition of sodium butyrate-induced apoptosis in recombinant Chinese hamster ovary cells by constitutively expressing antisense RNA of caspase-3”, Biotechn. & Bioengin. 78(2):217-228, 2002.
  • Knight et al., “Construction and initial characterization of a mouse-human chimeric anti-TNF antibody” (1993) Mol. Immunol., 30(16):1443-1453.
  • Kopaciewicz et al., “Retention Model for High-Performance Ion-Exchange Chromatography,”; Journal of Chromatography, 266:3-21 (1983).
  • Lerner, “Antibodies without immunization” (1992) Science, 258:1313-1314.
  • Leusch, “Failure to demonstrate TNFα-specific autoantibodies in human sera by ELISA and Western blot” (1991) J. Immunol. Methods, 139:145-47.
  • Lewis, “Use of alanine scanning mutagenesis to improve the affinity of an anti gp120 (HIV) antibody” (1994) J. Cell. Biochem., 18D:215.
  • Li, F. et al., “Current Therapeutic Antibody Production and Process Optimization” BioProcessing Journal, vol. 4(5):23-30 (2005).
  • Lifely et al., “Glycosylation and Biological Activity of CAMPATH-1H Expressed in Different Cell Lines and Grown Under Different Culture Conditions”, Glycobiology, 5(8):813-822 (1995).
  • Logan, John S. “Transgenic Animals: Beyond ‘Funny Milk’”, Current Opinion in Biotechnology, 4:591-595 (1993).
  • Lonberg et al., “Antigen-specific human antibodies from mice comprising four distinct genetic modifications” (1994) Nature, 368:856-859.
  • Lonberg et al., “Human Antibodies from Transgenic Mice” (1995) Int. Rev. Immunol., 13:65-93.
  • Low, “Mimicking Somatic Hypermutation: Affinity Maturation of Antibodies Displayed on Bacteriophage Using a Bacterial Mutator Strain” (1996) J. Mol. Biol., 260:359-368.
  • Low, Nigel: thesis extract (1996) Cambridge University.
  • Luo et al., “Understanding of C-terminal lysine variants in antibody production using mammalian cells” Abstract of papers, ACS, Anaheim, CA, US, Mar. 2011.
  • Luo et al: “Probing of C-terminal lysine variation in a recombinant monoclonal antibody production using Chinese hamster ovary cells with chemically defined media”, Biotechnology and Bioengineering, vol. 109, No. 9, Apr. 11, 2012, pp. 2306-2315.
  • Ma, et al., “Generation and Assembly of Secretory Antibodies in Plants”, Science, 268:716-719 (1995).
  • Maeda, et al., “Analysis of Nonhuman N-Glycans as the Minor Constituents in Recombinant Monoclonal Antibody Pharmaceuticals”, Anal. Chem., 84:2373-2379 (2012).
  • Mahler, et al. Induction and analysis of aggregates in a liquid IgG1-antibody formulation. Eur J Pharm Biopharm. 2005, 59(3):407-17; p. 408; col. 1-2; p. 409; col. 2, “2.2.2 Stirring stress”.
  • Marks et al., “Human antibody fragments specific for human blood group antigens from a phage display library” (1993) Bio/Technology, 11:1145-1150.
  • Marks et al., “Molecular evolution of proteins on filamentous phage. Mimicking the strategy of the immune system” (1992) J. Biol. Chem. 267:16007-16010.
  • Marks, “By-passing immunization: Human antibodies from V-gene libraries displayed on phage” (1991) J. Mol. Biol., 222:581-597.
  • Marks, “Human Monoclonal Antibodies from V-gene Repertoires Expressed on Bacteriophage.” In Antibody Engineering, Second Edition, edited by Carl A.K. Borrebaeck (1995), pp. 53-88. New York: Oxford Univ. Press.
  • Marks, JD., “By-Passing Immunization: Building High Affinity Human Antibodies by Chain Shuffling” (1992) Biotechnology, 10:779-783.
  • Martin, A.C.R. “Accessing the Kabat antibody sequence database by computer” (1996) Proteins: Structure, Function and Genetics, 25:130-133.
  • Medynski, “Phage Display: All Dressed Up and Ready to Role” (1994) Bio/Technology, 12:1134-1136.
  • Mendez et al., “Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice” (1997) Nature Genetics, 15:146-156.
  • Meuwly, F. et al., “Conversion of a CHO cell culture process from perfusion to fed-batch technology without altering product quality”, J.Biotechn. 123:106-116, 2006.
  • Miller et al. “Characterization of site-specific glycation during process development of a human therapeutic monoclonal antibody” Journal of Pharmaceutical Sciences, vol. 100, No. 7, Jul. 2011, 2543-2550.
  • Millipore, “Pellicon 2 Filters and Holders,” 2003, pp. 1-8.
  • Moore, A., et al., “Effects of temperature shift on cell cycle, apoptosis and nucleotide pools in CHO cell batch cultures”, Cytotechnology, 23:47-54, 1997.
  • Möller, Monoclonal antibodies to human tumor necrosis factor α: in vitro and vivo application (1990) Cytokine, 2(3):162-69.
  • Neuberger M. et al., “Mice perform a human repertoire” (1997) Nature, 386:25-26.
  • Ngo et al., “Kosmotropes enhance the yield of antibody purified by affinity chromatography using immobilized bacterial immunoglobulin binding proteins,” Journal of Immunoassay & Immunochemistry, (2008) 29(1):105-115.
  • Nilsson, “Antibody engineering” (1995) Current Opinion in Structural Biology, 5:450-456.
  • Nogal, B., Chhiba, K. and Emery, J. C. (2012), Select host cell proteins coelute with monoclonal antibodies in protein a chromatography. Biotechnol Progress, 28: 454-458.
  • Noguchi et al., “Failure of Human Immunoresponse to N-Glycolylneuraminic Acid Epitope Contained in Recombinant Human Erythropoietin”, Nephron, 72:599-603 (1996).
  • Noguchi et al., “Immunogenicity of N-Glycolylneuraminic Acid-Containing Carbohydrate Chains of Recombinant Human Erythropoietin Expressed in Chinese Hamster Ovary Cells”, J. Biochem., 117:59-62 (1995).
  • Oh, D-K. et al., “Increased erythritol production in fed-batch cultures of Torula sp. by controlling glucose concentration”, J. Industrial Microb. & Biotechn. 26(4): 248-252, 2001.
  • Oh, SKW, et al., “Substantial Overproduction of Antibodies by Applying Osmotic Pressure and Sodium Butyrate”, Biotechn. Bioengin. 42(5):601-610, 1993.
  • Osbourn, “From rodent reagents to human therapeutics using antibody guided selection” (2005) Methods, 36(1):61-68.
  • Perchiacca et al., “Aggregation-resistance domain antibodies engineered with charged mutations; near the edges of the complementarity-determining regions,” Protein Engineering Design & Selection, 25:10 (591-601) 2012.
  • Pietersz et al., “In vitro and in vivo Antitumor Activity of a Chimeric anti-CD19 Antibody”, Cancer Immunol. Immunother., 41:53-60 (1995).
  • Potter et al., “Antibody Production in the Baculovirus Expression System”, Intern. Rev. Immunol., 10:103-112 (1993).
  • Poul et al., “Design of Cassette Baculovirus Vectors for the Production of Therapeutic Antibodies in Insect Cells”, Immunotechnology, 1:189-196 (1995).
  • Queen, C., “A humanized antibody that binds to the interleukin 2 receptor” (1989) Proc. Natl. Acad. Sci. USA, 86(24):10029-10033.
  • Rader et al. “A phage display approach to rapid antibody humanization: Designed combinatorial V gene libraries” (1998) Proc Natl Acad Sci USA, 95:8910-8915.
  • Raju, TS. “Glycosylation Variations with Expression Systems and Their Impact on Biological Activity of Therapeutic Immunoglobulins”, BioProcess International., 44-53 (2003).
  • Reichert JM., et al., “Monoclonal antibody successes in the clinic”, Nature Biotech. 23(9):1073-1078, 2005.
  • Reinhart, “Assessment of the safety and efficacy of the monoclonal anti-tumor necrosis factor antibody-fragment, MAK 195F, in patients with sepsis and septic shock: a multicenter, randomized, placebo-controlled, dose-ranging study” (1996) Crit. Care Med., 24(5):733-742.
  • Rheinwald JG, et al., “Growth of Cultured Mammalian Cells on Secondary Glucose Sources”, Cell, 287-293, 1974.
  • Ridder et al., “Generation of Rabbit Monoclonal Antibody Fragments from a Combinatorial Phage Display Library and Their Production in Yeast Pichia pastoris”, Biotechnology, 13:255-260 (1995).
  • Riechmann, “Phage display and selection of a site-directed randomized single-chain antibody Fv fragment for its affinity improvement” (1993) Biochemistry, 32(34):8848-8855.
  • Rube et al. (Int. J. Radiation Oncology Biol. Phys., vol. 56, No. 5, pp. 1414-1425,2003).
  • Rudikoff et al., “Single amino acid substitution altering antigen-binding specificity” (1982) Proc. Natl. Acad. Sci. USA, 70:1979-1983.
  • Salfeld, “Development of a Fully Human Antibody to TNF by Phage Display Technology,” IBC Conference, Antibody Engineering, San Diego (Dec. 1996), pp. 1-36.
  • Sandadi, S. et al., “Heuristic Optimization of Antibody Production by Chinese Hamster Ovary Cells”, Biotech. Progress, American Institute of Chem. Engineers: 21(5): 1537-1542, 2005.
  • Sandhu, J. “Protein engineering of antibodies” (1992) Critical Reviews in Biotechnology, 12:437-462.
  • Santora et al., “Characterization of recombinant human monoclonal tissue necrosis factor-alpha antibody using cation exchange HPLC and capillary isoelectric focusing,” Analytical Biochemistry, (1999) 275:98-108.
  • Santora, “Characterization of Noncovalent Complexes of Recombinant Human Monoclonal Antibody and Antigen Using Cation Exchange, Size Exclusion Chromatography, and BIAcore” (2001) Analytical Biochemistry, 299:119-129.
  • Sato et al, “Stimulation of monoclonal antibody production by human-human hybridoma cells with an elevated concentration of potassium or sodium phosphate in serum-free medium,” Cytotechnology 2:63-67, 1989.
  • Schwieterman, “Immunosuppression in Combination with Monoclonal Antibodies” in Biologic Agents in Autoimmune Disease (Mar. 2-4, 1995).
  • Seresht et al., “The impact of phosphate scarcity on pharmaceutical protein production in S. cerevisiae: linking transcriptomic insights to phenotypic responses” Microbial Cell Factories. 2011, 10: 104.
  • Sheeley et al., “Characterization of Monoclonal Antibody Glycosylation: Comparison of Expression Systems and Identification of Terminal α-Linked Galactose”, Anal. Biochem., 247(1):102-110 (1997).
  • Sheikh et al., “Studies of the digestion of bradykinin, lysyl bradykinin, and kinin-degradation products by carboxypeptidases A, B, and N;”. Biochemical Pharmacology. 1986, 35: 1957-1963.
  • Shih, “Effects of Anions on the Deamidation of Soy Protein”. Journal of Food Science. 1991, 56: 452-454.
  • Shukla et al., “Host cell protein clearance during protein A chromatography: development of an improved column wash step,” Biotechnology Progress, (2008) 24(5):1115-1121.
  • Shukla et al., “Recent advances in large-scale production of monoclonal antibodies and related proteins,” Trends in Biotechnology, (2010) 28(5):253-261.
  • Sioud et al., “Characterization of naturally occurring autoantibodies against tumour necrosis factor-alpha (TNF-α): in vitro function and precise epitope mapping by phage epitope library” (1994) Clin. Exp. Immunol., 98:520-525.
  • Sung, Y.H. et al., “Yeast hydrolysate as a low-cost additive to serum-free medium for the production of human thrombpoietin in suspension cultures of Chinese hamster ovary cells”, Applied Microbilolgy and Biotechnology 63:5, 527-536, 2004.
  • Takagi, M. et al., “The effect of osmolarity on metabolism and morphology in adhesion and suspension chinese hamster ovary cells producing tissue plasminogen activator”, Cytochnology 32:171-179, 2000.
  • Tamura et al., “Structural correlates of an anticarcinoma antibody: identification of specificity-determining residues (SDRs) and development of a minimally immunogenic antibody variant by retention of SDFs only,” J. Immun. (2000) 164:1432-1441.
  • Tan et al. (Biotechnol. Appl. Biochem. (1999) 30, 59-64).
  • Taylor et al.,“Human immunoglobulin transgenes undergo rearrangement, somatic mutation and class switching in mice that lack endogenous IgM” (1994) Int. Immunol., 6:579-591.
  • Teichmann, S. Declaration dated Dec. 7, 2010 from opposition proceedings in EP 0929578.
  • The Kennedy Institute of Rheumatology, 1995 Annual Scientific Report, “Anti-TNF trials and studies of mechanisms of action”.
  • The MW Calculator available at the Sequence Manipulation Suite (see http://bioinformatics.org/sms2/index.html), downloaded Feb. 25, 2014.
  • The pl Calculator available at the Sequence Manipulation Suite (see <http://bioinformatics.org/sms2/index.html>), downloaded Feb. 25, 2014, p. 1).
  • The Statement on a Nonproprietary Name Adopted by the USAN Council for Adalimumab, p. 1, downloaded on May 19, 2011 from http://www.ama-assn.org/resources/doc/usan/adalimumab.doc.
  • Thompson, “Affinity maturation of a high-affinity human monoclonal antibody against the third hypervariable loop of human immunodeficiency virus: use of phage display to improve affinity and broaden strain reactivity” (1996) J. Mol. Biol., 256(1):77-88.
  • Thorp, “Tumour Necrosis Factor Induction of ELAM-1 and ICAM-1 on Human Umbilical Vein Endothelial Cells—Analysis of Tumour Necrosis Factor Receptor Interaction” (1992) Cytokine, 4(4): 313-319.
  • Tomiya et al., “Comparing N-glycan processing in mammalian cell lines to native and engineered; lepidopteran insect cell lines,” Glycoconjuqate Journal 21 :343-360 (2004).
  • Tomlinson, “The repertoire of human germline VH sequences reveals about fifty groups of VH segments with different hypervariable loops” (1992) J. Mol. Biol., 227:776-98.
  • Tomlinson, “The structural repertoire of the human Vk domain” (1995) The EMBO J., 14(18):4628-38.
  • Tracey, “Tumor necrosis factor: A pleiotropic cytokine and therapeutic target” (1994) Annu. Rev. Med., 45:491-503.
  • Tsuchiyama et al., “Comparison of anti-TNF alpha autoantibodies in plasma and from EBV transformed lymphocytes of autoimmune and normal individuals” (1995) Hum. Antibod. Hybridomas, 6(2):73-76.
  • Vallee B et al. “The role of zinc in carboxypeptidase” The Journal of Biological Chemistry, (1960) 235, 1; 64-69.
  • Valliere-Douglass et al., “Glutamine-linked and Non-consensus Asparagine-linked Oligosaccharides Present in Human Recombinant Antibodies Define Novel Protein Glycosylation Motifs”, J. Biol. Chem., 285:1 6012-16022 (2010).
  • Van Der Poll, “Effect of postponed treatment with an anti-tumour necrosis factor (TNF) F(ab′)2 fragment on endotoxin-induced cytokine and neutrophil responses in chimpanzees” (1995) Clin. Exp. Immunol., 100:21-25.
  • Van Lent PL, et al. “The impact of protein size and charge on its retention in articular cartilage” J Rheumatol. Aug. 1987;14(4):798-805.
  • Varasteh et al. Optimization of Anti-Rh D Immunoglobulin Stability in the Lyphiliization Process. Iranian Journal of Basic Medical Sciences, Spring 2008, vol. 11, No. 1. pp. 55-61.
  • Vaughan, “Human antibodies by design” (1998) Nature Biotechnology, 16:535-539.
  • Wagner et al., “Antibodies generated from human immunoglobulin miniloci in transgenic mice” (1994) Nucl. Acids Res. 22:1389-1393.
  • Wagner et al., “The diversity of antigen-specific monoclonal antibodies from transgenic mice bearing human immunoglobulin gene miniloci” (1994) Eur. J. Immunol., 24:2672-2681.
  • Ward, “Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli” (1989) Nature, 341:544-546.
  • Wedemayer et al., “Structural insights into the evolution of an antibody combining site” (1997) Science, 276:1665-1669.
  • Wiendl et al. (BioDrugs. 2002;16(3):183-200).
  • Williams et al., “Kinetic analysis by stopped-flow radiationless energy transfer studies: effect of anions on the activity of carboxypeptidase A”. Biochemistry. 1986, 25, 94-100.
  • Winter, “Humanized antibodies” (1993) Immunol. Today, 14(6):243-246.
  • Winter, “Making antibodies by phage display technology” (1994) Annu. Rev. Immunol., 12:433-455.
  • Wurm, FM, “Production of recombinant protein therapeutics in cultivated mammalian cells”, Nature Biotechnology 22(11):1393-1398, 2004.
  • Yigzaw et al., “Exploitation of the adsorptive properties of depth filters for host cell protein removal during monoclonal antibody purification,” Biotechnology Progress, (2006) 22(1):288-296.
  • Yumioka et al., “Screening of effective column rinse solvent for Protein-A chromatography,” Protein Expression and Purification, (2010) 70(2): 218-223.
  • Zatarain-Rios E and Mannik M, “Charge-charge interactions between articular cartilage and cationic antibodies, antigens, and immune complexes,” Arthritis Rheum. Nov. 1987;30(11):1265-73.
  • Zhang et al., “Isolation and characterization of charge variants using cation exchange displacement chromatography,” 1218(31): 5079-5086, 2011.
  • Zou et al., “Dominant expression of a 1.3 Mb human Ig kappa locus replacing mouse light chain production” (1996) FASEB J., 10:1227-1232.
Patent History
Patent number: 8921526
Type: Grant
Filed: Mar 12, 2014
Date of Patent: Dec 30, 2014
Patent Publication Number: 20140271626
Assignee: AbbVie, Inc. (North Chicago, IL)
Inventors: Chris M. Chumsae (North Andover, MA), Zehra Kaymakcalan (Westborough, MA)
Primary Examiner: David Romeo
Application Number: 14/206,296